151
|
Sam E, Athri P. Web-based drug repurposing tools: a survey. Brief Bioinform 2017; 20:299-316. [DOI: 10.1093/bib/bbx125] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Indexed: 12/15/2022] Open
Affiliation(s)
- Elizabeth Sam
- Department of Computer Science & Engineering Amrita, University Bengaluru, India
| | - Prashanth Athri
- Department of Computer Science & Engineering Amrita, University Bengaluru, India
| |
Collapse
|
152
|
Tanaka M, Yoshino Y, Takeda S, Toda K, Shimoda H, Tsuruma K, Shimazawa M, Hara H. Fermented Rice Germ Extract Alleviates Morphological and Functional Damage to Murine Gastrocnemius Muscle by Inactivation of AMP-Activated Protein Kinase. J Med Food 2017; 20:969-980. [PMID: 28956710 DOI: 10.1089/jmf.2016.3906] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Sarcopenia, loss of muscle mass and function, is mainly observed in elderly people. In this study, we investigated whether fermented rice germ extract (FRGE) has some effects on the mouse gastrocnemius muscle by using behavioral and morphological analyses, Western blotting, and a murine model of immobilization-induced muscle atrophy. Daily oral FRGE administration increased muscle weight and strength. In addition, myofiber size in gastrocnemius muscle of FRGE-treated mice was increased as revealed by morphological quantification. Activation of AMP-activated protein kinase (AMPK) signaling, which inhibits protein synthesis and stimulates protein degradation in gastrocnemius muscle, was significantly attenuated in the FRGE-treated mice compared with control mice. Expression level of forkhead box 3a (FOXO3a) protein was also significantly decreased in the FRGE-treated group. Moreover, the decrease in mean myofiber cross-sectional area in immobilized hindlimb in vehicle-treated mice was inhibited by FRGE treatment in histological analysis. In conclusion, FRGE increased the strength and weight of gastrocnemius muscle and myofiber size, and reduced immobilization-induced muscle atrophy in mice. These findings indicated that FRGE might be beneficial in preventing motor dysfunction in a range of conditions, including sarcopenia.
Collapse
Affiliation(s)
- Miyu Tanaka
- 1 Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University , Gifu, Japan
| | - Yuta Yoshino
- 1 Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University , Gifu, Japan
| | - Shogo Takeda
- 2 Oryza Oil & Fat Chemical Co., Ltd. , Ichinomiya, Japan
| | - Kazuya Toda
- 2 Oryza Oil & Fat Chemical Co., Ltd. , Ichinomiya, Japan
| | | | - Kazuhiro Tsuruma
- 1 Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University , Gifu, Japan
| | - Masamitsu Shimazawa
- 1 Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University , Gifu, Japan
| | - Hideaki Hara
- 1 Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University , Gifu, Japan
| |
Collapse
|
153
|
Jang YJ, Son HJ, Choi YM, Ahn J, Jung CH, Ha TY. Apigenin enhances skeletal muscle hypertrophy and myoblast differentiation by regulating Prmt7. Oncotarget 2017; 8:78300-78311. [PMID: 29108230 PMCID: PMC5667963 DOI: 10.18632/oncotarget.20962] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 08/27/2017] [Indexed: 12/19/2022] Open
Abstract
Apigenin, a natural flavone abundant in various plant-derived foods including parsley and celery, has been shown to prevent inflammation and inflammatory diseases. However, the effect of apigenin on skeletal muscle hypertrophy and myogenic differentiation has not previously been elucidated. Here, we investigated the effects of apigenin on quadricep muscle weight and running distance using C57BL/6 mice on an accelerating treadmill. Apigenin stimulated mRNA expression of MHC (myosin heavy chain) 1, MHC2A, and MHC2B in the quadricep muscles of these animals. GPR56 (G protein-coupled receptor 56) and its ligand collagen III were upregulated by apigenin supplementation, together with enhanced PGC-1α, PGC-1α1, PGC-1α4, IGF1, and IGF2 expression. Prmt7 protein expression increased in conjunction with Akt and mTORC1 activation. Apigenin treatment also upregulated FNDC5 (fibronectin type III domain containing 5) mRNA expression and serum irisin levels. Furthermore, apigenin stimulated C2C12 myogenic differentiation and upregulated total MHC, MHC2A, and MHC2B expression. These events were attributable to an increase in Prmt7-p38-myoD expression and Akt and S6K1 phosphorylation. We also observed that Prmt7 regulates both PGC-1α1 and PGC-1α4 expression, resulting in a subsequent increase in GPR56 expression and mTORC1 activation. Taken together, these findings suggest that apigenin supplementation can promote skeletal muscle hypertrophy and myogenic differentiation by regulating the Prmt7-PGC-1α-GPR56 pathway, as well as the Prmt7-p38-myoD pathway, which may contribute toward the prevention of skeletal muscle weakness.
Collapse
Affiliation(s)
- Young Jin Jang
- Division of Nutrition and Metabolism Research, Korea Food Research Institute, Seongnam, Republic of Korea
| | - Hyo Jeong Son
- Division of Nutrition and Metabolism Research, Korea Food Research Institute, Seongnam, Republic of Korea
| | - Yong Min Choi
- Division of Nutrition and Metabolism Research, Korea Food Research Institute, Seongnam, Republic of Korea
| | - Jiyun Ahn
- Division of Nutrition and Metabolism Research, Korea Food Research Institute, Seongnam, Republic of Korea.,Division of Food Biotechnology, University of Science and Technology, Daejeon, Republic of Korea
| | - Chang Hwa Jung
- Division of Nutrition and Metabolism Research, Korea Food Research Institute, Seongnam, Republic of Korea.,Division of Food Biotechnology, University of Science and Technology, Daejeon, Republic of Korea
| | - Tae Youl Ha
- Division of Nutrition and Metabolism Research, Korea Food Research Institute, Seongnam, Republic of Korea.,Division of Food Biotechnology, University of Science and Technology, Daejeon, Republic of Korea
| |
Collapse
|
154
|
Rodriguez J, Pierre N, Naslain D, Bontemps F, Ferreira D, Priem F, Deldicque L, Francaux M. Urolithin B, a newly identified regulator of skeletal muscle mass. J Cachexia Sarcopenia Muscle 2017; 8:583-597. [PMID: 28251839 PMCID: PMC5566634 DOI: 10.1002/jcsm.12190] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 01/01/2017] [Accepted: 01/10/2017] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND The control of muscle size is an essential feature of health. Indeed, skeletal muscle atrophy leads to reduced strength, poor quality of life, and metabolic disturbances. Consequently, strategies aiming to attenuate muscle wasting and to promote muscle growth during various (pathological) physiological states like sarcopenia, immobilization, malnutrition, or cachexia are needed to address this extensive health issue. In this study, we tested the effects of urolithin B, an ellagitannin-derived metabolite, on skeletal muscle growth. METHODS C2C12 myotubes were treated with 15 μM of urolithin B for 24 h. For in vivo experiments, mice were implanted with mini-osmotic pumps delivering continuously 10 μg/day of urolithin B during 28 days. Muscle atrophy was studied in mice with a sciatic nerve denervation receiving urolithin B by the same way. RESULTS Our experiments reveal that urolithin B enhances the growth and differentiation of C2C12 myotubes by increasing protein synthesis and repressing the ubiquitin-proteasome pathway. Genetic and pharmacological arguments support an implication of the androgen receptor. Signalling analyses suggest a crosstalk between the androgen receptor and the mTORC1 pathway, possibly via AMPK. In vivo experiments confirm that urolithin B induces muscle hypertrophy in mice and reduces muscle atrophy after the sciatic nerve section. CONCLUSIONS This study highlights the potential usefulness of urolithin B for the treatment of muscle mass loss associated with various (pathological) physiological states.
Collapse
Affiliation(s)
- Julie Rodriguez
- Institute of Neuroscience, Université catholique de Louvain, 1 place Pierre de Coubertin, 1348, Louvain-la-Neuve, Belgium.,PROCELL nutrition sprl, 2 Rue Jean Burgers, 7850, Enghien, Belgium
| | - Nicolas Pierre
- Institute of Neuroscience, Université catholique de Louvain, 1 place Pierre de Coubertin, 1348, Louvain-la-Neuve, Belgium
| | - Damien Naslain
- Institute of Neuroscience, Université catholique de Louvain, 1 place Pierre de Coubertin, 1348, Louvain-la-Neuve, Belgium
| | - Françoise Bontemps
- De Duve Institute, Université catholique de Louvain, 75 Avenue Hippocrate, 1200, Brussels, Belgium
| | - Daneel Ferreira
- Department of Biomolecular Sciences, Division of Pharmacognosy, Research Institute of Pharmaceutical Sciences, University of Mississippi, Medicinal Plant Garden, RM 104, University, MS, 38677, USA
| | - Fabian Priem
- PROCELL nutrition sprl, 2 Rue Jean Burgers, 7850, Enghien, Belgium
| | - Louise Deldicque
- Institute of Neuroscience, Université catholique de Louvain, 1 place Pierre de Coubertin, 1348, Louvain-la-Neuve, Belgium
| | - Marc Francaux
- Institute of Neuroscience, Université catholique de Louvain, 1 place Pierre de Coubertin, 1348, Louvain-la-Neuve, Belgium
| |
Collapse
|
155
|
Chen J, Wong HS, Leong PK, Leung HY, Chan WM, Ko KM. Ursolic acid induces mitochondrial biogenesis through the activation of AMPK and PGC-1 in C2C12 myotubes: a possible mechanism underlying its beneficial effect on exercise endurance. Food Funct 2017; 8:2425-2436. [PMID: 28675237 DOI: 10.1039/c7fo00127d] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Mitochondrial biogenesis, which involves an increase in mitochondrial number and the overall capacity of oxidative phosphorylation, is a critical determinant of skeletal muscle function. Recent findings have shown that some natural products can enhance mitochondrial adaptation to aerobic exercise, which in turn improves exercise performance, presumably by delaying muscle fatigue. Ursolic acid (UA), a natural triterpene, is commonly found in various vegetables and fruits. In the current study, UA was shown to increase mitochondrial mass and ATP generation capacity, with a concomitant production of a low level of mitochondrial reactive oxygen species (ROS) in C2C12 myotubes. Mitochondrial ROS, in turn, activated the redox sensitive adenosine monophosphate-dependent protein kinase (AMPK)/peroxisome proliferator-activated receptor γ coactivator-1(PGC-1) pathway. The activation of AMPK/PGC-1 further increased the expression of cytochrome c oxidase (COX) and uncoupling protein 3. Animal studies showed that UA can also dose-dependently increase the endurance exercise capacity in mice, as assessed by a weight-loaded swimming test and a hanging wire test. Our findings suggest that UA may induce mitochondrial biogenesis through the activation of AMPK and PGC-1 pathways in skeletal muscle, thereby offering a promising prospect for its use to enhance exercise endurance and alleviating fatigue in humans.
Collapse
Affiliation(s)
- Jihang Chen
- Division of Life Science, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong SAR, China.
| | | | | | | | | | | |
Collapse
|
156
|
Vilar S, Hripcsak G. The role of drug profiles as similarity metrics: applications to repurposing, adverse effects detection and drug-drug interactions. Brief Bioinform 2017; 18:670-681. [PMID: 27273288 PMCID: PMC6078166 DOI: 10.1093/bib/bbw048] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 04/18/2016] [Indexed: 12/30/2022] Open
Abstract
Explosion of the availability of big data sources along with the development in computational methods provides a useful framework to study drugs' actions, such as interactions with pharmacological targets and off-targets. Databases related to protein interactions, adverse effects and genomic profiles are available to be used for the construction of computational models. In this article, we focus on the description of biological profiles for drugs that can be used as a system to compare similarity and create methods to predict and analyze drugs' actions. We highlight profiles constructed with different biological data, such as target-protein interactions, gene expression measurements, adverse effects and disease profiles. We focus on the discovery of new targets or pathways for drugs already in the pharmaceutical market, also called drug repurposing, in the interaction with off-targets responsible for adverse reactions and in drug-drug interaction analysis. The current and future applications, strengths and challenges facing all these methods are also discussed. Biological profiles or signatures are an important source of data generation to deeply analyze biological actions with important implications in drug-related studies.
Collapse
Affiliation(s)
- Santiago Vilar
- Corresponding author: Santiago Vilar, Department of Biomedical Informatics, Columbia University Medical Center, New York, NY 10032, USA. E-mail: ; George Hripcsak, Department of Biomedical Informatics, Columbia University Medical Center, New York, NY 10032, USA. E-mail:
| | - George Hripcsak
- Corresponding author: Santiago Vilar, Department of Biomedical Informatics, Columbia University Medical Center, New York, NY 10032, USA. E-mail: ; George Hripcsak, Department of Biomedical Informatics, Columbia University Medical Center, New York, NY 10032, USA. E-mail:
| |
Collapse
|
157
|
Katashima CK, Silva VR, Gomes TL, Pichard C, Pimentel GD. Ursolic acid and mechanisms of actions on adipose and muscle tissue: a systematic review. Obes Rev 2017; 18:700-711. [PMID: 28335087 DOI: 10.1111/obr.12523] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Revised: 01/21/2017] [Accepted: 01/22/2017] [Indexed: 12/28/2022]
Abstract
This systematic review aimed at addressing the ursolic acid actions as an adjunctive treatment of the obesity-mediated metabolic abnormalities. To explore our aims, we used the literature search including clinical and animal studies using the Medline and Google Scholar (up to December 2015). Out of 63 screened studies, 17 presented eligibility criteria, such as the use of ursolic acid on adiposity, energy expenditure and skeletal muscle mass in mice and humans. In the literature, we found that several physiological and molecular mechanisms are implicated in the effects of ursolic acid on obesity, energy expenditure, hepatic steatosis, skeletal muscle mass loss and physical fitness, such as (1) increase of thermogenesis by modulation adipocyte transcription factors, activation of 5' adenosine monophosphate-activated protein kinase and overexpression of the uncoupling protein 1 thermogenic marker; (2) enhancement of skeletal muscle mass by activation in bloodstream growth hormone and insulin-like growth factor-1 concentrations secretion, as well as in the activation of mammalian target of rapamycin and inhibition of ring-finger protein-1; and (3) improvement of physical fitness by skeletal muscle proliferator-activated receptor gamma co-activator alpha and sirtuin 1 expression. Therefore, supplementation with ursolic acid may be an adjunctive therapy for prevention and treatment of obesity-mediated and muscle mass-mediated metabolic consequences.
Collapse
Affiliation(s)
| | | | - Tatyanne L Gomes
- Clinical and Sports Nutrition Research Laboratory (Labince), School of Nutrition (FANUT), Federal University of Goias (UFG), Goiânia, GO, Brazil
| | - Claude Pichard
- Nutrition Unit, Geneva University Hospital, Geneva, Switzerland
| | - Gustavo D Pimentel
- Clinical and Sports Nutrition Research Laboratory (Labince), School of Nutrition (FANUT), Federal University of Goias (UFG), Goiânia, GO, Brazil
| |
Collapse
|
158
|
Mining the transcriptome for rare disease therapies: a comparison of the efficiencies of two data mining approaches and a targeted cell-based drug screen. NPJ Genom Med 2017; 2:14. [PMID: 29263829 PMCID: PMC5677978 DOI: 10.1038/s41525-017-0018-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 03/15/2017] [Accepted: 03/17/2017] [Indexed: 01/18/2023] Open
Abstract
Most monogenic diseases can be viewed as conditions caused by dysregulated protein activity; therefore, drugs can be used to modulate gene expression, and thus protein level, possibly conferring clinical benefit. When considering repurposing drugs for loss of function diseases, there are three classes of genetic disease amenable to an increase of function; haploinsufficient dominant diseases, those secondary to hypomorphic recessive alleles, and conditions with rescuing paralogs. This therapeutic model then brings the questions: how frequently do such clinically useful drug–gene interactions occur and what is the most rapid and efficient route by which to identify them. Here we compare three approaches: (1) mining of pre-existing system-wide transcriptomal datasets such as Connectivity Map; (2) utilization of a proprietary causal reasoning engine knowledge base; and, (3) a targeted drug screen using clinically accepted agents tested against normal human fibroblasts. We have determined the validation rate of these approaches for 76 diseases (i.e., in vitro fibroblast mRNA increase); for the Connectivity Map, approximately 5% of tested putative drug–gene interactions validated, for causal reasoning engine knowledge base the rate was 10%, and for the targeted drug screen 9%. The degree of overlap between these methodologies was low suggesting they are complementary not redundant approaches to identify putative drug-gene interactions. Although the validation rate was low, a number of drug–gene interactions were successfully identified and are now being investigated for protein induction and in vivo effect. This analysis establishes potentially valuable therapeutic leads as well as useful benchmarks for the thousands of currently untreatable rare genetic conditions. Canadian researchers use three different approaches to identify drugs that could be repurposed to treat rare genetic diseases. Prompted by the growing gap between known rare diseases and therapies and postulating that diseases might be treated with drugs that modulate levels of disease-related proteins, Alex Mackenzie at the University of Ottawa and colleagues, studied the impact of 300 clinically approved drugs on 75 rare disease associated genes. Analyses of Connectivity Map gene expression data, published texts and a cell-based screening assay revealed thousands of putative and potentially clinically beneficial interactions. Although just 5–10% altered protein expression, the different approaches were complementary rather than redundant methodologies identifying potentially useful starting points on the path to rare disease therapy.
Collapse
|
159
|
Deane CS, Wilkinson DJ, Phillips BE, Smith K, Etheridge T, Atherton PJ. "Nutraceuticals" in relation to human skeletal muscle and exercise. Am J Physiol Endocrinol Metab 2017; 312:E282-E299. [PMID: 28143855 PMCID: PMC5406990 DOI: 10.1152/ajpendo.00230.2016] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 01/25/2017] [Accepted: 01/25/2017] [Indexed: 12/16/2022]
Abstract
Skeletal muscles have a fundamental role in locomotion and whole body metabolism, with muscle mass and quality being linked to improved health and even lifespan. Optimizing nutrition in combination with exercise is considered an established, effective ergogenic practice for athletic performance. Importantly, exercise and nutritional approaches also remain arguably the most effective countermeasure for muscle dysfunction associated with aging and numerous clinical conditions, e.g., cancer cachexia, COPD, and organ failure, via engendering favorable adaptations such as increased muscle mass and oxidative capacity. Therefore, it is important to consider the effects of established and novel effectors of muscle mass, function, and metabolism in relation to nutrition and exercise. To address this gap, in this review, we detail existing evidence surrounding the efficacy of a nonexhaustive list of macronutrient, micronutrient, and "nutraceutical" compounds alone and in combination with exercise in relation to skeletal muscle mass, metabolism (protein and fuel), and exercise performance (i.e., strength and endurance capacity). It has long been established that macronutrients have specific roles and impact upon protein metabolism and exercise performance, (i.e., protein positively influences muscle mass and protein metabolism), whereas carbohydrate and fat intakes can influence fuel metabolism and exercise performance. Regarding novel nutraceuticals, we show that the following ones in particular may have effects in relation to 1) muscle mass/protein metabolism: leucine, hydroxyl β-methylbutyrate, creatine, vitamin-D, ursolic acid, and phosphatidic acid; and 2) exercise performance: (i.e., strength or endurance capacity): hydroxyl β-methylbutyrate, carnitine, creatine, nitrates, and β-alanine.
Collapse
Affiliation(s)
- Colleen S Deane
- Medical Research Council-Arthritis Research UK Centre of Excellence for Musculoskeletal Ageing Research and Clinical, Metabolic, and Molecular Physiology, University of Nottingham, Royal Derby Hospital, Derby, United Kingdom
- Faculty of Health and Social Science, Bournemouth University, Bournemouth, United Kingdom; and
- Department of Sport and Health Science, College of Life and Environmental Sciences, University of Exeter, Exeter, United Kingdom
| | - Daniel J Wilkinson
- Medical Research Council-Arthritis Research UK Centre of Excellence for Musculoskeletal Ageing Research and Clinical, Metabolic, and Molecular Physiology, University of Nottingham, Royal Derby Hospital, Derby, United Kingdom
| | - Bethan E Phillips
- Medical Research Council-Arthritis Research UK Centre of Excellence for Musculoskeletal Ageing Research and Clinical, Metabolic, and Molecular Physiology, University of Nottingham, Royal Derby Hospital, Derby, United Kingdom
| | - Kenneth Smith
- Medical Research Council-Arthritis Research UK Centre of Excellence for Musculoskeletal Ageing Research and Clinical, Metabolic, and Molecular Physiology, University of Nottingham, Royal Derby Hospital, Derby, United Kingdom
| | - Timothy Etheridge
- Department of Sport and Health Science, College of Life and Environmental Sciences, University of Exeter, Exeter, United Kingdom
| | - Philip J Atherton
- Medical Research Council-Arthritis Research UK Centre of Excellence for Musculoskeletal Ageing Research and Clinical, Metabolic, and Molecular Physiology, University of Nottingham, Royal Derby Hospital, Derby, United Kingdom;
| |
Collapse
|
160
|
Misra RC, Sharma S, Garg A, Chanotiya CS, Ghosh S. Two CYP716A subfamily cytochrome P450 monooxygenases of sweet basil play similar but nonredundant roles in ursane- and oleanane-type pentacyclic triterpene biosynthesis. THE NEW PHYTOLOGIST 2017; 214:706-720. [PMID: 28967669 DOI: 10.1111/nph.14412] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 11/23/2016] [Indexed: 05/23/2023]
Abstract
The medicinal plant sweet basil (Ocimum basilicum) accumulates bioactive ursane- and oleanane-type pentacyclic triterpenes (PCTs), ursolic acid and oleanolic acid, respectively, in a spatio-temporal manner; however, the biosynthetic enzymes and their contributions towards PCT biosynthesis remain to be elucidated. Two CYP716A subfamily cytochrome P450 monooxygenases (CYP716A252 and CYP716A253) are identified from a methyl jasmonate-responsive expression sequence tag collection and functionally characterized, employing yeast (Saccharomyces cerevisiae) expression platform and adapting virus-induced gene silencing (VIGS) in sweet basil. CYP716A252 and CYP716A253 catalyzed sequential three-step oxidation at the C-28 position of α-amyrin and β-amyrin to produce ursolic acid and oleanolic acid, respectively. Although CYP716A253 was more efficient than CYP716A252 for amyrin C-28 oxidation in yeast, VIGS revealed essential roles for both of these CYP716As in constitutive biosynthesis of ursolic acid and oleanolic acid in sweet basil leaves. However, CYP716A253 played a major role in elicitor-induced biosynthesis of ursolic acid and oleanolic acid. Overall, the results suggest similar as well as distinct roles of CYP716A252 and CYP716A253 for the spatio-temporal biosynthesis of PCTs. CYP716A252 and CYP716A253 might be useful for the alternative and sustainable production of PCTs in microbial host, besides increasing plant metabolite content through genetic modification.
Collapse
Affiliation(s)
- Rajesh Chandra Misra
- Biotechnology Division, Council of Scientific and Industrial Research-Central Institute of Medicinal and Aromatic Plants, Lucknow, 226015, India
| | - Shubha Sharma
- Biotechnology Division, Council of Scientific and Industrial Research-Central Institute of Medicinal and Aromatic Plants, Lucknow, 226015, India
- Academy of Scientific and Innovative Research, New Delhi, India
| | - Anchal Garg
- Biotechnology Division, Council of Scientific and Industrial Research-Central Institute of Medicinal and Aromatic Plants, Lucknow, 226015, India
| | - Chandan Singh Chanotiya
- Analytical Chemistry Division, Council of Scientific and Industrial Research-Central Institute of Medicinal and Aromatic Plants, Lucknow, 226015, India
| | - Sumit Ghosh
- Biotechnology Division, Council of Scientific and Industrial Research-Central Institute of Medicinal and Aromatic Plants, Lucknow, 226015, India
- Academy of Scientific and Innovative Research, New Delhi, India
| |
Collapse
|
161
|
Yu R, Chen J, Xu J, Cao J, Wang Y, Thomas SS, Hu Z. Suppression of muscle wasting by the plant-derived compound ursolic acid in a model of chronic kidney disease. J Cachexia Sarcopenia Muscle 2017; 8:327-341. [PMID: 27897418 PMCID: PMC5377392 DOI: 10.1002/jcsm.12162] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 09/16/2016] [Accepted: 09/27/2016] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Muscle wasting in chronic kidney disease (CKD) and other catabolic disorders contributes to morbidity and mortality, and there are no therapeutic interventions that regularly and safely block losses of muscle mass. We have obtained evidence that impaired IGF-1/insulin signalling and increases in glucocorticoids, myostatin and/or inflammatory cytokines that contribute to the development of muscle wasting in catabolic disorders by activating protein degradation. METHODS Using in vitro and in vivo models of muscle wasting associated with CKD or dexamethasone administration, we measured protein synthesis and degradation and examined mechanisms by which ursolic acid, derived from plants, could block the loss of muscle mass stimulated by CKD or excessive levels of dexamethasone. RESULTS Using cultured C2C12 myotubes to study muscle wasting, we found that exposure to glucocorticoids cause loss of cell proteins plus an increase in myostatin; both responses are significantly suppressed by ursolic acid. Results from promoter and ChIP assays demonstrated a mechanism involving ursolic acid blockade of myostatin promoter activity that is related to CEBP/δ expression. In mouse models of CKD-induced or dexamethasone-induced muscle wasting, we found that ursolic acid blocked the loss of muscle mass by stimulating protein synthesis and decreasing protein degradation. These beneficial responses included decreased expression of myostatin and inflammatory cytokines (e.g. TGF-β, IL-6 and TNFα), which are initiators of muscle-specific ubiquitin-E3 ligases (e.g. Atrogin-1, MuRF-1 and MUSA1). CONCLUSIONS Ursolic acid improves CKD-induced muscle mass by suppressing the expression of myostatin and inflammatory cytokines via increasing protein synthesis and reducing proteolysis.
Collapse
Affiliation(s)
- Rizhen Yu
- Nephrology DivisionChanghai HospitalShanghaiChina
- Nephrology DivisionZhejiang Provincial People's HospitalHongzhouChina
- Nephrology DivisionDepartment of Medicine, Baylor College of MedicineHoustonTXUSA
| | - Ji‐an Chen
- Department of Health Education, College of Preventive MedicineThird Military Medical UniversityChongqingChina
| | - Jing Xu
- Nephrology DivisionChanghai HospitalShanghaiChina
| | - Jin Cao
- Nephrology DivisionDepartment of Medicine, Baylor College of MedicineHoustonTXUSA
| | - Yanlin Wang
- Nephrology DivisionDepartment of Medicine, Baylor College of MedicineHoustonTXUSA
| | - Sandhya S. Thomas
- Michael E. Debakey VA Medical CenterHoustonTXUSA
- Nephrology DivisionDepartment of Medicine, Baylor College of MedicineHoustonTXUSA
| | - Zhaoyong Hu
- Nephrology DivisionDepartment of Medicine, Baylor College of MedicineHoustonTXUSA
| |
Collapse
|
162
|
Jeong J, Park CH, Kim I, Kim YH, Yoon JM, Kim KS, Kim JB. Korean mistletoe (Viscum album coloratum) extract regulates gene expression related to muscle atrophy and muscle hypertrophy. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2017; 17:68. [PMID: 28109285 PMCID: PMC5251312 DOI: 10.1186/s12906-017-1575-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/18/2016] [Accepted: 01/11/2017] [Indexed: 02/05/2023]
Abstract
Background Korean mistletoe (Viscum album coloratum) is a semi-parasitic plant that grows on various trees and has a diverse range of effects on biological functions, being implicated in having anti-tumor, immunostimulatory, anti-diabetic, and anti-obesity properties. Recently, we also reported that Korean mistletoe extract (KME) improves endurance exercise in mice, suggesting its beneficial roles in enhancing the capacity of skeletal muscle. Methods We examined the expression pattern of several genes concerned with muscle physiology in C2C12 myotubes cells to identify whether KME inhibits muscle atrophy or promotes muscle hypertrophy. We also investigated these effects of KME in denervated mice model. Results Interestingly, KME induced the mRNA expression of SREBP-1c, PGC-1α, and GLUT4, known positive regulators of muscle hypertrophy, in C2C12 cells. On the contrary, KME reduced the expression of Atrogin-1, which is directly involved in the induction of muscle atrophy. In animal models, KME mitigated the decrease of muscle weight in denervated mice. The expression of Atrogin-1 was also diminished in those mice. Moreover, KME enhanced the grip strength and muscle weight in long-term feeding mice. Conclusions Our results suggest that KME has beneficial effects on muscle atrophy and muscle hypertrophy.
Collapse
|
163
|
De Gasperi R, Graham ZA, Harlow LM, Bauman WA, Qin W, Cardozo CP. The Signature of MicroRNA Dysregulation in Muscle Paralyzed by Spinal Cord Injury Includes Downregulation of MicroRNAs that Target Myostatin Signaling. PLoS One 2016; 11:e0166189. [PMID: 27907012 PMCID: PMC5132212 DOI: 10.1371/journal.pone.0166189] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 10/24/2016] [Indexed: 12/25/2022] Open
Abstract
Spinal cord injury (SCI) results in muscle atrophy, reduced force generation and an oxidative-to-glycolytic fiber type shift. The mechanisms responsible for these alterations remain incompletely understood. To gain new insights regarding mechanisms involved in deterioration of muscle after SCI, global expression profiles of miRs in paralyzed gastrocnemius muscle were compared between sham-operated (Sham) and spinal cord-transected (SCI) rats. Ingenuity Pathways Analysis of the altered miRs identified signaling via insulin, IGF-1, integrins and TGF-β as being significantly enriched for target genes. By qPCR, miRs 23a, 23b, 27b, 145, and 206, were downregulated in skeletal muscle 56 days after SCI. Using FISH, miR-145, a miR not previously implicated in the function of skeletal muscle, was found to be localized to skeletal muscle fibers. One predicted target of miR-145 was Cited2, a transcriptional regulator that modulates signaling through NF-κB, Smad3 and other transcription factors. The 3’ UTR of Cited2 mRNA contained a highly conserved miR-145 seed sequence. Luciferase reporter assays confirmed that miR-145 interacts with this seed sequence. However, Cited2 protein levels were similar between Sham and SCI groups, indicating a biochemical interaction that was not involved in the context of adaptations after SCI. Taken together, the findings indicate dysregulation of several highly expressed miRs in skeletal muscle after SCI and suggest that reduced expression of miR-23a, 145 and 206 may have roles in alteration in skeletal muscle mass and insulin responsiveness in muscle paralyzed by upper motor neuron injuries.
Collapse
Affiliation(s)
- Rita De Gasperi
- VA RR&D Service National Center for the Medical Consequences of Spinal Cord Injury, James J. Peters Medical Center, Bronx, New York
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Zachary A. Graham
- VA RR&D Service National Center for the Medical Consequences of Spinal Cord Injury, James J. Peters Medical Center, Bronx, New York
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Lauren M. Harlow
- VA RR&D Service National Center for the Medical Consequences of Spinal Cord Injury, James J. Peters Medical Center, Bronx, New York
| | - William A. Bauman
- VA RR&D Service National Center for the Medical Consequences of Spinal Cord Injury, James J. Peters Medical Center, Bronx, New York
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Rehabilitation Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Weiping Qin
- VA RR&D Service National Center for the Medical Consequences of Spinal Cord Injury, James J. Peters Medical Center, Bronx, New York
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Christopher P. Cardozo
- VA RR&D Service National Center for the Medical Consequences of Spinal Cord Injury, James J. Peters Medical Center, Bronx, New York
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Rehabilitation Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Pharmacologic Science Icahn School of Medicine at Mount Sinai, New York, New York
- * E-mail:
| |
Collapse
|
164
|
Cho YH, Lee SY, Kim CM, Kim ND, Choe S, Lee CH, Shin JH. Effect of Loquat Leaf Extract on Muscle Strength, Muscle Mass, and Muscle Function in Healthy Adults: A Randomized, Double-Blinded, and Placebo-Controlled Trial. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2016; 2016:4301621. [PMID: 27999607 PMCID: PMC5143716 DOI: 10.1155/2016/4301621] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Revised: 07/17/2016] [Accepted: 08/16/2016] [Indexed: 12/02/2022]
Abstract
Ursolic acid (UA) is the major active component of the loquat leaf extract (LLE) and several previous studies have indicated that UA may have the ability to prevent skeletal muscle atrophy. Therefore, we conducted a randomized, double-blind, and placebo-controlled study to investigate the effects of the LLE on muscle strength, muscle mass, muscle function, and metabolic markers in healthy adults; the safety of the compound was also evaluated. We examined the peak torque/body weight at 60°/s knee extension, handgrip strength, skeletal muscle mass, physical performance, and metabolic parameters at baseline, as well as after 4 and 12 weeks of intervention. Either 500 mg of LLE (50.94 mg of UA) or a placebo was administered to fifty-four healthy adults each day for 12 weeks; no differences in muscle strength, muscle mass, and physical performance were observed between the two groups. However, the right-handgrip strength of female subjects in the LLE group was found to be significantly better than that of subjects in the control group (P = 0.047). Further studies are required to determine the optimal dose and duration of LLE supplementation to confirm the first-stage study results for clinical application. ClinicalTrials.gov Identifier is NCT02401113.
Collapse
Affiliation(s)
- Young Hye Cho
- Family Medicine Clinic, Obesity, Metabolism and Nutrition Center and Research Institute of Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| | - Sang Yeoup Lee
- Family Medicine Clinic, Obesity, Metabolism and Nutrition Center and Research Institute of Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
- Department of Medical Education, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Cheol Min Kim
- Center for Anti-Aging Industry, Pusan National University, Busan, Republic of Korea
- Department of Biomedical Informatics, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Nam Deuk Kim
- Department of Pharmacy, Molecular Inflammation Research Center for Aging Intervention, Pusan National University, Busan, Republic of Korea
| | - Sangmin Choe
- Department of Clinical Pharmacology and Therapeutics, Pusan National University Hospital, Busan, Republic of Korea
| | - Chang-Hyung Lee
- Department of Rehabilitation Medicine, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| | - Jin-Hong Shin
- Department of Neurology, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| |
Collapse
|
165
|
Yoon Y, Lim JW, Kim J, Kim Y, Chun KH. Discovery of ursolic acid prodrug (NX-201): Pharmacokinetics and in vivo antitumor effects in PANC-1 pancreatic cancer. Bioorg Med Chem Lett 2016; 26:5524-5527. [PMID: 27769622 DOI: 10.1016/j.bmcl.2016.10.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 09/22/2016] [Accepted: 10/05/2016] [Indexed: 01/09/2023]
Abstract
The aim of our study was to develop ursolic acid (UA) prodrugs in order to overcome UA's weakness, which has an extremely low bioavailability. UA-medoxomil (NX-201), one of our UA prodrugs, showed an improved bioavailability about 200times better than UA in rodent model. According to in vivo test performed with PANC-1 xenograft SCID mouse model, tumor growth rate decreased dose-dependently and 100mg/kg dose of NX-201 had an anticancer effect comparable to gemcitabine. Most of all the combination of NX-201 (50mg/kg, po, daily) and gemcitabine (40mg/kg, iv, 2timesperweek) even reduced tumor size after three weeks.
Collapse
Affiliation(s)
- Yeohong Yoon
- Nexoligo Co., Ltd., 40 4F 3408, Simin-daero 365 beongil, Dongan-gu, Anyang-si, Gyeonggi-do 14057, Republic of Korea; Department of Chemistry, Soongsil University, 369, Sangdo-ro, Dongjak-gu, Seoul 06978, Republic of Korea
| | - Jee Woong Lim
- Nexoligo Co., Ltd., 40 4F 3408, Simin-daero 365 beongil, Dongan-gu, Anyang-si, Gyeonggi-do 14057, Republic of Korea
| | - Jiyoung Kim
- Nexoligo Co., Ltd., 40 4F 3408, Simin-daero 365 beongil, Dongan-gu, Anyang-si, Gyeonggi-do 14057, Republic of Korea
| | - Younggi Kim
- KNOTUS Co., Ltd., 189, Dongureung-ro, Guri-si, Gyeonggi-do 02117, Republic of Korea
| | - Keun Ho Chun
- Department of Chemistry, Soongsil University, 369, Sangdo-ro, Dongjak-gu, Seoul 06978, Republic of Korea
| |
Collapse
|
166
|
Saul MC, Majdak P, Perez S, Reilly M, Garland T, Rhodes JS. High motivation for exercise is associated with altered chromatin regulators of monoamine receptor gene expression in the striatum of selectively bred mice. GENES BRAIN AND BEHAVIOR 2016; 16:328-341. [DOI: 10.1111/gbb.12347] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Revised: 10/02/2016] [Accepted: 10/03/2016] [Indexed: 01/12/2023]
Affiliation(s)
- M. C. Saul
- Carl R. Woese Institute for Genomic Biology Urbana IL
| | | | - S. Perez
- The Beckman Institute for Advanced Science and Technology University of Illinois Urbana IL
| | - M. Reilly
- National Institute on Alcohol Abuse and Alcoholism National Institutes of Health Bethesda MD
| | - T. Garland
- Department of Biology University of California Riverside CA
| | - J. S. Rhodes
- Carl R. Woese Institute for Genomic Biology Urbana IL
- The Neuroscience Program
- The Beckman Institute for Advanced Science and Technology University of Illinois Urbana IL
- Department of Psychology University of Illinois Urbana IL USA
| |
Collapse
|
167
|
Moro T, Ebert SM, Adams CM, Rasmussen BB. Amino Acid Sensing in Skeletal Muscle. Trends Endocrinol Metab 2016; 27:796-806. [PMID: 27444066 PMCID: PMC5075248 DOI: 10.1016/j.tem.2016.06.010] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Revised: 06/23/2016] [Accepted: 06/27/2016] [Indexed: 12/19/2022]
Abstract
Aging impairs skeletal muscle protein synthesis, leading to muscle weakness and atrophy. However, the underlying molecular mechanisms remain poorly understood. Here, we review evidence that mammalian/mechanistic target of rapamycin complex 1 (mTORC1)-mediated and activating transcription factor 4 (ATF4)-mediated amino acid (AA) sensing pathways, triggered by impaired AA delivery to aged skeletal muscle, may play important roles in skeletal muscle aging. Interventions that alleviate age-related impairments in muscle protein synthesis, strength, and/or muscle mass appear to do so by reversing age-related changes in skeletal muscle AA delivery, mTORC1 activity, and/or ATF4 activity. An improved understanding of the mechanisms and roles of AA sensing pathways in skeletal muscle may lead to evidence-based strategies to attenuate sarcopenia.
Collapse
Affiliation(s)
- Tatiana Moro
- Department of Nutrition and Metabolism, University of Texas Medical Branch, Galveston, TX, USA; Sealy Center on Aging, University of Texas Medical Branch, Galveston, TX, USA
| | - Scott M Ebert
- Department of Internal Medicine, University of Iowa, Iowa City, IA, USA; Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, USA; Iowa City Veterans Affairs Medical Center, Iowa City, IA, USA
| | - Christopher M Adams
- Department of Internal Medicine, University of Iowa, Iowa City, IA, USA; Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, USA; Iowa City Veterans Affairs Medical Center, Iowa City, IA, USA
| | - Blake B Rasmussen
- Department of Nutrition and Metabolism, University of Texas Medical Branch, Galveston, TX, USA; Sealy Center on Aging, University of Texas Medical Branch, Galveston, TX, USA.
| |
Collapse
|
168
|
Ma P, Yao L, Lin X, Gu T, Rong X, Batey R, Yamahara J, Wang J, Li Y. A mixture of apple pomace and rosemary extract improves fructose consumption-induced insulin resistance in rats: modulation of sarcolemmal CD36 and glucose transporter-4. Am J Transl Res 2016; 8:3791-3801. [PMID: 27725859 PMCID: PMC5040677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 08/19/2016] [Indexed: 06/06/2023]
Abstract
Apple pomace is a by-product of the processing of apple for juice, cider or wine preparation. Rosemary is a herb commonly used as spice and flavoring agent in food processing. Evidence suggests that both apple pomace and rosemary have rich bioactive molecules with numerous metabolic effects. To provide more information for using apple pomace and rosemary as functional foods for management of metabolism-associated disorders, the present study investigated the insulin-sensitizing effect of a mixture of apple pomace and rosemary extract (AR). The results showed that treatment with AR (500 mg/kg, daily, by gavage) for 5 weeks attenuated chronic liquid fructose consumption-induced increases in fasting plasma insulin concentration, the homeostasis model assessment of insulin resistance index and the adipose tissue insulin resistance index in rats. Mechanistically, AR suppressed fructose-induced acceleration of the clearance of plasma non-esterified fatty acids during oral glucose tolerance test, and decreased excessive triglyceride accumulation and the increased Oil Red O staining area in the gastrocnemius. Furthermore, AR restored fructose-induced overexpression of sarcolemmal CD36 that is known to contribute to etiology of insulin resistance by facilitating fatty acid uptake, and downregulation of sarcolemmal glucose transporter (GLUT)-4 that is the insulin-responsive glucose transporter. Thus, these results demonstrate that AR improves fructose-induced insulin resistance in rats via modulation of sarcolemmal CD36 and GLUT-4.
Collapse
Affiliation(s)
- Peng Ma
- Faculty of Basic Medical Sciences, Chongqing Medical UniversityChongqing 400016, China
| | - Ling Yao
- The Laboratory of Traditional Chinese Medicine, Chongqing Medical UniversityChongqing 400016, China
| | - Xuemei Lin
- Faculty of Basic Medical Sciences, Chongqing Medical UniversityChongqing 400016, China
| | - Tieguang Gu
- Endocrinology and Metabolism Group, Sydney Institute of Health Sciences/Sydney Institute of Traditional Chinese MedicineNSW 2000, Australia
| | - Xianglu Rong
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, and The Institute of Chinese Medicine, Guangdong Pharmaceutical UniversityChina
| | - Robert Batey
- Central Clinical School, Royal Prince Alfred Hospital, The University of SydneyAustralia
| | | | - Jianwei Wang
- The Laboratory of Traditional Chinese Medicine, Chongqing Medical UniversityChongqing 400016, China
| | - Yuhao Li
- Endocrinology and Metabolism Group, Sydney Institute of Health Sciences/Sydney Institute of Traditional Chinese MedicineNSW 2000, Australia
| |
Collapse
|
169
|
Church DD, Schwarz NA, Spillane MB, McKinley-Barnard SK, Andre TL, Ramirez AJ, Willoughby DS. l-Leucine Increases Skeletal Muscle IGF-1 but Does Not Differentially Increase Akt/mTORC1 Signaling and Serum IGF-1 Compared to Ursolic Acid in Response to Resistance Exercise in Resistance-Trained Men. J Am Coll Nutr 2016; 35:627-638. [PMID: 27331824 DOI: 10.1080/07315724.2015.1132019] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
OBJECTIVE Ursolic acid administration following resistance exercise increases mammalian target of rapamycin complex 1 (mTORC1) activity and skeletal muscle IGF-1 concentration in murines in a manner similar to l-leucine yet remains unexamined in humans. This study examined serum and skeletal muscle insulin-like growth factor-1 (IGF-1) and Akt/mTORC1 signaling activity following ingestion of either ursolic acid or l-leucine immediately after resistance exercise. METHODS Nine resistance-trained men performed 3 lower-body resistance exercise sessions involving 4 sets of 8-10 repetitions at 75%-80% one repetition maximum (1-RM) on the angled leg press and knee extension exercises. Immediately following each session, participants orally ingested 3 g cellulose placebo (PLC), l-leucine (LEU), or ursolic acid (UA). Blood samples were obtained pre-exercise and at 0.5, 2, and 6 hours postexercise. Muscle biopsies were obtained pre-exercise and at 2 and 6 hours postexercise. RESULTS Plasma leucine increased in LEU at 2 hours postexercise compared to PLC (p = 0.04). Plasma ursolic acid increased in UA at 2 h and 6 hours postexercise compared to PLC and LEU (p < 0.003). No significant differences were observed for serum insulin (p = 0.98) and IGF-1 (p = 0.99) or skeletal muscle IGF-1 receptor (IGF-1R; p = 0.84), Akt (p = 0.55), mTOR (p = 0.09), and p70S6K (p = 0.98). Skeletal muscle IGF-1 was significantly increased in LEU at 2 hours postexercise (p = 0.03) and 6 hours postexercise (p = 0.04) compared to PLC and UA. CONCLUSION Three grams of l-leucine and ursolic acid had no effect on Akt/mTORC1 signaling or serum insulin or IGF-1; however, l-leucine increased skeletal muscle IGF-1 concentration in resistance-trained men.
Collapse
Affiliation(s)
- David D Church
- a Exercise and Biochemical Nutrition Lab, Department of Health , Human Performance, and Recreation, College of Arts and Sciences, Baylor University , Waco , Texas
| | - Neil A Schwarz
- a Exercise and Biochemical Nutrition Lab, Department of Health , Human Performance, and Recreation, College of Arts and Sciences, Baylor University , Waco , Texas
| | - Mike B Spillane
- a Exercise and Biochemical Nutrition Lab, Department of Health , Human Performance, and Recreation, College of Arts and Sciences, Baylor University , Waco , Texas
| | - Sarah K McKinley-Barnard
- a Exercise and Biochemical Nutrition Lab, Department of Health , Human Performance, and Recreation, College of Arts and Sciences, Baylor University , Waco , Texas
| | - Tom L Andre
- a Exercise and Biochemical Nutrition Lab, Department of Health , Human Performance, and Recreation, College of Arts and Sciences, Baylor University , Waco , Texas
| | - Alejandro J Ramirez
- b Mass Spectrometry Center , College of Arts and Sciences, Baylor University , Waco , Texas
| | - Darryn S Willoughby
- a Exercise and Biochemical Nutrition Lab, Department of Health , Human Performance, and Recreation, College of Arts and Sciences, Baylor University , Waco , Texas
| |
Collapse
|
170
|
Rom O, Reznick AZ. The role of E3 ubiquitin-ligases MuRF-1 and MAFbx in loss of skeletal muscle mass. Free Radic Biol Med 2016; 98:218-230. [PMID: 26738803 DOI: 10.1016/j.freeradbiomed.2015.12.031] [Citation(s) in RCA: 138] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 11/30/2015] [Accepted: 12/25/2015] [Indexed: 12/21/2022]
Abstract
The ubiquitin-proteasome system (UPS) is the main regulatory mechanism of protein degradation in skeletal muscle. The ubiquitin-ligase enzymes (E3s) have a central role in determining the selectivity and specificity of the UPS. Since their identification in 2001, the muscle specific E3s, muscle RING finger-1 (MuRF-1) and muscle atrophy F-box (MAFbx), have been shown to be implicated in the regulation of skeletal muscle atrophy in various pathological and physiological conditions. This review aims to explore the involvement of MuRF-1 and MAFbx in catabolism of skeletal muscle during various pathologies, such as cancer cachexia, sarcopenia of aging, chronic kidney disease (CKD), diabetes, and chronic obstructive pulmonary disease (COPD). In addition, the effects of various lifestyle and modifiable factors (e.g. nutrition, exercise, cigarette smoking, and alcohol) on MuRF-1 and MAFbx regulation will be discussed. Finally, evidence of potential strategies to protect against skeletal muscle wasting through inhibition of MuRF-1 and MAFbx expression will be explored.
Collapse
Affiliation(s)
- Oren Rom
- Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, P.O. Box 9649, Haifa, Israel.
| | - Abraham Z Reznick
- Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, P.O. Box 9649, Haifa, Israel
| |
Collapse
|
171
|
Close GL, Hamilton DL, Philp A, Burke LM, Morton JP. New strategies in sport nutrition to increase exercise performance. Free Radic Biol Med 2016; 98:144-158. [PMID: 26855422 DOI: 10.1016/j.freeradbiomed.2016.01.016] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 01/19/2016] [Accepted: 01/21/2016] [Indexed: 02/03/2023]
Abstract
Despite over 50 years of research, the field of sports nutrition continues to grow at a rapid rate. Whilst the traditional research focus was one that centred on strategies to maximise competition performance, emerging data in the last decade has demonstrated how both macronutrient and micronutrient availability can play a prominent role in regulating those cell signalling pathways that modulate skeletal muscle adaptations to endurance and resistance training. Nonetheless, in the context of exercise performance, it is clear that carbohydrate (but not fat) still remains king and that carefully chosen ergogenic aids (e.g. caffeine, creatine, sodium bicarbonate, beta-alanine, nitrates) can all promote performance in the correct exercise setting. In relation to exercise training, however, it is now thought that strategic periods of reduced carbohydrate and elevated dietary protein intake may enhance training adaptations whereas high carbohydrate availability and antioxidant supplementation may actually attenuate training adaptation. Emerging evidence also suggests that vitamin D may play a regulatory role in muscle regeneration and subsequent hypertrophy following damaging forms of exercise. Finally, novel compounds (albeit largely examined in rodent models) such as epicatechins, nicotinamide riboside, resveratrol, β-hydroxy β-methylbutyrate, phosphatidic acid and ursolic acid may also promote or attenuate skeletal muscle adaptations to endurance and strength training. When taken together, it is clear that sports nutrition is very much at the heart of the Olympic motto, Citius, Altius, Fortius (faster, higher, stronger).
Collapse
Affiliation(s)
- G L Close
- Research Institute for Sport and Exercise Science (RISES), Liverpool John Moores University, Tom Reilly Building, Byrom Street, Liverpool L3 3AF, United Kingdom.
| | - D L Hamilton
- Health and Exercise Sciences Research Group, University of Stirling, Stirling, United Kingdom
| | - A Philp
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, United Kingdom
| | - L M Burke
- Sports Nutrition, Australian Institute of Sport, Canberra, ACT, Australia; Mary Mackillop Institute for Health Research, Melbourne, Australia
| | - J P Morton
- Research Institute for Sport and Exercise Science (RISES), Liverpool John Moores University, Tom Reilly Building, Byrom Street, Liverpool L3 3AF, United Kingdom
| |
Collapse
|
172
|
Bahrami SA, Bakhtiari N. Ursolic acid regulates aging process through enhancing of metabolic sensor proteins level. Biomed Pharmacother 2016; 82:8-14. [PMID: 27470332 DOI: 10.1016/j.biopha.2016.04.047] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2016] [Revised: 04/23/2016] [Accepted: 04/25/2016] [Indexed: 02/06/2023] Open
Abstract
We previously reported that Ursolic Acid (UA) ameliorates skeletal muscle performance through satellite cells proliferation and cellular energy status. In studying the potential role of the hypothalamus in aging, we developed a strategy to pursue UA effects on the hypothalamus anti-aging proteins such as; SIRT1, SIRT6, PGC-1β and α-Klotho. In this study, we used a model of aging animals (C57BL/6). UA dissolved in Corn oil (20mg/ml) and then administrated (200mg/Kg i.p injection) to mice, twice daily for 7days. After treatment times, the mice perfused and the hypothalamus isolated for preparing of tissue to Immunofluorescence microscopy. The data illustrated that UA significantly increased SIRT1 (∼3.5±0.3 folds) and SIRT-6 (∼1.5±0.2 folds) proteins overexpression (P<0.001). In addition, our results showed that UA enhanced α-Klotho (∼3.3±0.3) and PGC-1β (∼2.6±0.2 folds) proteins levels (P<0. 01). In this study, data were analyzed using SPSS 16 (ANOVA test). To the best of our knowledge, it seems that UA through enhancing of anti-aging biomarkers (SIRT1 and SIRT6) and PGC-1β in hypothalamus regulates aging-process and attenuates mitochondrial-related diseases. In regard to the key role of α-Klotho in aging, our data indicate that UA may be on the horizon to forestall diseases of aging.
Collapse
Affiliation(s)
- Soroush Alaghehband Bahrami
- Department of Biochemistry, Faculty of Basic Sciences, Sanandaj Branch, Islamic Azad University, Sanandaj, Iran
| | - Nuredin Bakhtiari
- Department of Biochemistry, Faculty of Basic Sciences, Sanandaj Branch, Islamic Azad University, Sanandaj, Iran.
| |
Collapse
|
173
|
Anthony TG. Mechanisms of protein balance in skeletal muscle. Domest Anim Endocrinol 2016; 56 Suppl:S23-32. [PMID: 27345321 PMCID: PMC4926040 DOI: 10.1016/j.domaniend.2016.02.012] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 02/23/2016] [Accepted: 02/29/2016] [Indexed: 01/07/2023]
Abstract
Increased global demand for adequate protein nutrition against a backdrop of climate change and concern for animal agriculture sustainability necessitates new and more efficient approaches to livestock growth and production. Anabolic growth is achieved when rates of new synthesis exceed turnover, producing a positive net protein balance. Conversely, deterioration or atrophy of lean mass is a consequence of a net negative protein balance. During early life and periods of growth, muscle mass is driven by increases in protein synthesis at the level of mRNA translation. Throughout life, muscle mass is further influenced by degradative processes such as autophagy and the ubiquitin proteasome pathway. Multiple signal transduction networks guide and coordinate these processes alongside quality control mechanisms to maintain protein homeostasis (proteostasis). Genetics, hormones, and environmental stimuli each influence proteostasis control, altering capacity and/or efficiency of muscle growth. An overview of recent findings and current methods to assess muscle protein balance and proteostasis is presented. Current efforts to identify novel control points have the potential through selective breeding design or development of hormetic strategies to better promote growth and health span during environmental stress.
Collapse
Affiliation(s)
- T G Anthony
- Department of Nutritional Sciences, Rutgers University, New Brunswick, NJ 08901, USA.
| |
Collapse
|
174
|
Vilar S, Hripcsak G. Leveraging 3D chemical similarity, target and phenotypic data in the identification of drug-protein and drug-adverse effect associations. J Cheminform 2016; 8:35. [PMID: 27375776 PMCID: PMC4930585 DOI: 10.1186/s13321-016-0147-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 06/23/2016] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Drug-target identification is crucial to discover novel applications for existing drugs and provide more insights about mechanisms of biological actions, such as adverse drug effects (ADEs). Computational methods along with the integration of current big data sources provide a useful framework for drug-target and drug-adverse effect discovery. RESULTS In this article, we propose a method based on the integration of 3D chemical similarity, target and adverse effect data to generate a drug-target-adverse effect predictor along with a simple leveraging system to improve identification of drug-targets and drug-adverse effects. In the first step, we generated a system for multiple drug-target identification based on the application of 3D drug similarity into a large target dataset extracted from the ChEMBL. Next, we developed a target-adverse effect predictor combining targets from ChEMBL with phenotypic information provided by SIDER data source. Both modules were linked to generate a final predictor that establishes hypothesis about new drug-target-adverse effect candidates. Additionally, we showed that leveraging drug-target candidates with phenotypic data is very useful to improve the identification of drug-targets. The integration of phenotypic data into drug-target candidates yielded up to twofold precision improvement. In the opposite direction, leveraging drug-phenotype candidates with target data also yielded a significant enhancement in the performance. CONCLUSIONS The modeling described in the current study is simple and efficient and has applications at large scale in drug repurposing and drug safety through the identification of mechanism of action of biological effects.
Collapse
Affiliation(s)
- Santiago Vilar
- Department of Biomedical Informatics, Columbia University Medical Center, New York, NY USA
| | - George Hripcsak
- Department of Biomedical Informatics, Columbia University Medical Center, New York, NY USA
| |
Collapse
|
175
|
Lin YT, Yu YM, Chang WC, Chiang SY, Chan HC, Lee MF. Ursolic acid plays a protective role in obesity-induced cardiovascular diseases. Can J Physiol Pharmacol 2016; 94:627-33. [PMID: 26991492 DOI: 10.1139/cjpp-2015-0407] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2023]
Abstract
The metabolic disturbance of obesity is one of the most common risk factors of atherosclerosis. Resistin, an obesity-induced adipokine, can induce the expression of cell adhesion molecules and the attachment of monocytes to endothelial cells, which play an important role in the development of atherosclerosis. Ursolic acid, a pentacyclic triterpenoid found in fruits and many herbs, exhibits an array of biological effects such as anti-inflammatory and antioxidative properties. The aim of this study was to investigate the potential underlying mechanisms of the effect of ursolic acid on resistin-induced adhesion of U937 cells to human umbilical vein endothelial cells (HUVECs). Our data indicated that ursolic acid suppressed the adhesion of U937 to HUVECs and downregulated the expression of adhesion molecules, vascular cell adhesion molecule-1 (VCAM-1), intracellular cell adhesion molecule-1 (ICAM-1), and E-selectin, in resistin-induced HUVECs by decreasing the production of intracellular reaction oxygen species (ROS) and attenuating the nuclear translocation of NFκB. Ursolic acid appeared to inhibit resistin-induced atherosclerosis, suggesting that ursolic acid may play a protective role in obesity-induced cardiovascular diseases.
Collapse
Affiliation(s)
- Yu-Ting Lin
- a Graduate Institute of Nutrition, China Medical University, Taichung, Taiwan
| | - Ya-Mei Yu
- b Department of Nutrition and Health Sciences, Chang Jung Christian University, Tainan, Taiwan
| | - Weng-Cheng Chang
- c Graduate Institute of Medical Science, Chang Jung Christian University, Tainan, Taiwan
| | - Su-Yin Chiang
- d Graduate Institute of Chinese Medical Science, China Medical University, Taichung, Taiwan
| | - Hsu-Chin Chan
- e Department of Biochemistry, China Medical University, Taichung, Taiwan
| | - Ming-Fen Lee
- b Department of Nutrition and Health Sciences, Chang Jung Christian University, Tainan, Taiwan
| |
Collapse
|
176
|
A gene-signature progression approach to identifying candidate small-molecule cancer therapeutics with connectivity mapping. BMC Bioinformatics 2016; 17:211. [PMID: 27170106 PMCID: PMC4864913 DOI: 10.1186/s12859-016-1066-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Accepted: 04/29/2016] [Indexed: 01/14/2023] Open
Abstract
Background Gene expression connectivity mapping has gained much popularity recently with a number of successful applications in biomedical research testifying its utility and promise. Previously methodological research in connectivity mapping mainly focused on two of the key components in the framework, namely, the reference gene expression profiles and the connectivity mapping algorithms. The other key component in this framework, the query gene signature, has been left to users to construct without much consensus on how this should be done, albeit it has been an issue most relevant to end users. As a key input to the connectivity mapping process, gene signature is crucially important in returning biologically meaningful and relevant results. This paper intends to formulate a standardized procedure for constructing high quality gene signatures from a user’s perspective. Results We describe a two-stage process for making quality gene signatures using gene expression data as initial inputs. First, a differential gene expression analysis comparing two distinct biological states; only the genes that have passed stringent statistical criteria are considered in the second stage of the process, which involves ranking genes based on statistical as well as biological significance. We introduce a “gene signature progression” method as a standard procedure in connectivity mapping. Starting from the highest ranked gene, we progressively determine the minimum length of the gene signature that allows connections to the reference profiles (drugs) being established with a preset target false discovery rate. We use a lung cancer dataset and a breast cancer dataset as two case studies to demonstrate how this standardized procedure works, and we show that highly relevant and interesting biological connections are returned. Of particular note is gefitinib, identified as among the candidate therapeutics in our lung cancer case study. Our gene signature was based on gene expression data from Taiwan female non-smoker lung cancer patients, while there is evidence from independent studies that gefitinib is highly effective in treating women, non-smoker or former light smoker, advanced non-small cell lung cancer patients of Asian origin. Conclusions In summary, we introduced a gene signature progression method into connectivity mapping, which enables a standardized procedure for constructing high quality gene signatures. This progression method is particularly useful when the number of differentially expressed genes identified is large, and when there is a need to prioritize them to be included in the query signature. The results from two case studies demonstrate that the approach we have developed is capable of obtaining pertinent candidate drugs with high precision. Electronic supplementary material The online version of this article (doi:10.1186/s12859-016-1066-x) contains supplementary material, which is available to authorized users.
Collapse
|
177
|
Wang K, Wan M, Wang RS, Weng Z. Opportunities for Web-based Drug Repositioning: Searching for Potential Antihypertensive Agents with Hypotension Adverse Events. J Med Internet Res 2016; 18:e76. [PMID: 27036325 PMCID: PMC4833875 DOI: 10.2196/jmir.4541] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Revised: 12/02/2015] [Accepted: 01/04/2016] [Indexed: 12/21/2022] Open
Abstract
Background Drug repositioning refers to the process of developing new indications for existing drugs. As a phenotypic indicator of drug response in humans, clinical side effects may provide straightforward signals and unique opportunities for drug repositioning. Objective We aimed to identify drugs frequently associated with hypotension adverse reactions (ie, the opposite condition of hypertension), which could be potential candidates as antihypertensive agents. Methods We systematically searched the electronic records of the US Food and Drug Administration (FDA) Adverse Event Reporting System (FAERS) through the openFDA platform to assess the association between hypotension incidence and antihypertensive therapeutic effect regarding a list of 683 drugs. Results Statistical analysis of FAERS data demonstrated that those drugs frequently co-occurring with hypotension events were more likely to have antihypertensive activity. Ranked by the statistical significance of frequent hypotension reporting, the well-known antihypertensive drugs were effectively distinguished from others (with an area under the receiver operating characteristic curve > 0.80 and a normalized discounted cumulative gain of 0.77). In addition, we found a series of antihypertensive agents (particularly drugs originally developed for treating nervous system diseases) among the drugs with top significant reporting, suggesting the good potential of Web-based and data-driven drug repositioning. Conclusions We found several candidate agents among the hypotension-related drugs on our list that may be redirected for lowering blood pressure. More important, we showed that a pharmacovigilance system could alternatively be used to identify antihypertensive agents and sustainably create opportunities for drug repositioning.
Collapse
Affiliation(s)
- Kejian Wang
- CoMed Technology & Consulting Co., Ltd., Hong Kong, China
| | | | | | | |
Collapse
|
178
|
Wagner A, Cohen N, Kelder T, Amit U, Liebman E, Steinberg DM, Radonjic M, Ruppin E. Drugs that reverse disease transcriptomic signatures are more effective in a mouse model of dyslipidemia. Mol Syst Biol 2016; 11:791. [PMID: 26148350 PMCID: PMC4380926 DOI: 10.15252/msb.20145486] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
High-throughput omics have proven invaluable in studying human disease, and yet day-to-day clinical practice still relies on physiological, non-omic markers. The metabolic syndrome, for example, is diagnosed and monitored by blood and urine indices such as blood cholesterol levels. Nevertheless, the association between the molecular and the physiological manifestations of the disease, especially in response to treatment, has not been investigated in a systematic manner. To this end, we studied a mouse model of diet-induced dyslipidemia and atherosclerosis that was subject to various drug treatments relevant to the disease in question. Both physiological data and gene expression data (from the liver and white adipose) were analyzed and compared. We find that treatments that restore gene expression patterns to their norm are associated with the successful restoration of physiological markers to their baselines. This holds in a tissue-specific manner—treatments that reverse the transcriptomic signatures of the disease in a particular tissue are associated with positive physiological effects in that tissue. Further, treatments that introduce large non-restorative gene expression alterations are associated with unfavorable physiological outcomes. These results provide a sound basis to in silico methods that rely on omic metrics for drug repurposing and drug discovery by searching for compounds that reverse a disease's omic signatures. Moreover, they highlight the need to develop drugs that restore the global cellular state to its healthy norm rather than rectify particular disease phenotypes.
Collapse
Affiliation(s)
- Allon Wagner
- The Blavatnik School of Computer Science, Tel Aviv UniversityTel Aviv, Israel
- Department of Electrical Engineering and Computer Science, University of CaliforniaBerkeley, CA, USA
- * Corresponding author. Tel. +972 3 640 5378; E-mail:
| | - Noa Cohen
- The Blavatnik School of Computer Science, Tel Aviv UniversityTel Aviv, Israel
| | - Thomas Kelder
- Microbiology and Systems Biology, TNOZeist, the Netherlands
| | - Uri Amit
- Neufeld Cardiac Research Institute, Tel Aviv UniversityTel Aviv, Israel
- Regenerative Medicine Stem Cells and Tissue Engineering Center, Sheba Medical CenterTel Hashomer, Israel
| | - Elad Liebman
- Department of Computer Science, University of Texas at AustinAustin, TX, USA
| | - David M Steinberg
- Department of Statistics and Operations Research, Tel Aviv UniversityTel Aviv, Israel
| | | | - Eytan Ruppin
- The Blavatnik School of Computer Science, Tel Aviv UniversityTel Aviv, Israel
- The Sackler School of Medicine, Tel Aviv UniversityTel Aviv, Israel
- Department of Computer Science, Institute of Advanced Computer Sciences (UMIACS) & the Center for Bioinformatics and Computational Biology, University of MarylandCollege Park, MD, USA
- ** Corresponding author. Tel. +972 3 640 6528; E-mail:
| |
Collapse
|
179
|
Bond P. Regulation of mTORC1 by growth factors, energy status, amino acids and mechanical stimuli at a glance. J Int Soc Sports Nutr 2016; 13:8. [PMID: 26937223 PMCID: PMC4774173 DOI: 10.1186/s12970-016-0118-y] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 02/18/2016] [Indexed: 12/05/2022] Open
Abstract
The mechanistic/mammalian target of rapamycin complex 1 (mTORC1) plays a pivotal role in the regulation of skeletal muscle protein synthesis. Activation of the complex leads to phosphorylation of two important sets of substrates, namely eIF4E binding proteins and ribosomal S6 kinases. Phosphorylation of these substrates then leads to an increase in protein synthesis, mainly by enhancing translation initiation. mTORC1 activity is regulated by several inputs, such as growth factors, energy status, amino acids and mechanical stimuli. Research in this field is rapidly evolving and unraveling how these inputs regulate the complex. Therefore this review attempts to provide a brief and up-to-date narrative on the regulation of this marvelous protein complex. Additionally, some sports supplements which have been shown to regulate mTORC1 activity are discussed.
Collapse
Affiliation(s)
- Peter Bond
- PeterBond.nl, Waterhoenlaan 25, Zeist, Netherlands
| |
Collapse
|
180
|
Bakhtiari N, Hosseinkhani S, Soleimani M, Hemmati R, Noori-Zadeh A, Javan M, Tashakor A. Short-term ursolic acid promotes skeletal muscle rejuvenation through enhancing of SIRT1 expression and satellite cells proliferation. Biomed Pharmacother 2016; 78:185-196. [PMID: 26898441 DOI: 10.1016/j.biopha.2016.01.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Revised: 01/09/2016] [Accepted: 01/13/2016] [Indexed: 10/22/2022] Open
Abstract
Ursolic acid (UA) is a triterpenoid compound, which exerts its influences on the skeletal muscles. However, the mechanisms underlying these effects are still unclear. In this study, muscle satellite cells were isolated and purified by high-throughput pre-plating method (∼>60%) from 10 days old mice skeletal muscles. Evaluation of paired-box 7 (Pax7) expressions then confirmed the purification. Treatment of the cells with UA showed that UA up-regulated SIRT1 (∼35 folds) and overexpressed PGC-1α (∼175 folds) gene significantly. Moreover, the number of muscle satellite cells, which accompanied by initiation of neomyogenesis in the animal skeletal muscles, was increased (∼3.4 times). We also evaluated UA-mediated changes in the cellular energy status in the skeletal muscles. The results revealed that in the UA-treated mice, ATP and ADP contents in the various skeletal muscle tissue types, including: Gastrocnemius (Gas), Tibialis Anterior (Tib) and Gluteus Maximus (Glu) have been significantly decreased (P≤0.001); 2.2, 3.2, 2 times for ATP, and 9.6, 35.7, 11.6 times for ADP, respectively; however to compensate this process mitochondrial biogenesis occurred (12.33%±1.5 times). Furthermore, a rise in ATP/ADP ratio was observed 2.5, 4.5, 2.05 times for Gas, Tib and Glu muscles, respectively (P≤0.001). Alternatively, UA enhanced the expression of myoglobin (∼2 folds) in concert with remodeling of glycolytic muscle fibers to mainly fast IIA (∼30%) and slow-twitch (∼4%) types as well. Finally, our study indicated that UA indirectly mimicked beneficial effects of short-term calorie restriction and exercise (fast-oxidative) by directing the skeletal muscle composition toward oxidative metabolism.
Collapse
MESH Headings
- Adenosine Diphosphate/metabolism
- Adenosine Triphosphate/metabolism
- Animals
- Blood Glucose/metabolism
- Cell Line
- Cell Proliferation/drug effects
- Cell Separation
- Energy Metabolism/drug effects
- Fasting/blood
- Male
- Mice, Inbred C57BL
- Muscle Fibers, Fast-Twitch/drug effects
- Muscle Fibers, Fast-Twitch/metabolism
- Muscle Fibers, Slow-Twitch/drug effects
- Muscle Fibers, Slow-Twitch/metabolism
- Muscle, Skeletal/drug effects
- Muscle, Skeletal/physiology
- Myoglobin/metabolism
- Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism
- Rejuvenation/physiology
- Satellite Cells, Skeletal Muscle/cytology
- Satellite Cells, Skeletal Muscle/drug effects
- Satellite Cells, Skeletal Muscle/metabolism
- Satellite Cells, Skeletal Muscle/ultrastructure
- Sirtuin 1/metabolism
- Triterpenes/pharmacology
- Ursolic Acid
Collapse
Affiliation(s)
- Nuredin Bakhtiari
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Saman Hosseinkhani
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Masoud Soleimani
- Department of Hematology, School of Medical Sciences, Tarbiat Modares University, Tehran, Iran; Department of Stem Cell Biology, Stem Cell Technology Research Center, Tehran, Iran
| | - Roohullah Hemmati
- Department of Biology, Faculty of Basic Sciences, Shahrekord University, Iran
| | - Ali Noori-Zadeh
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mohammad Javan
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Amin Tashakor
- Institute of Biochemistry and Biophysics, Tehran University, Tehran, Iran
| |
Collapse
|
181
|
Sato H, Funaki A, Kimura Y, Sumitomo M, Yoshida H, Fukata H, Ueno K. Ethanol extract of Cyclolepis genistoides D. Don (palo azul) induces formation of myotubes, which involves differentiation of C2C12 myoblast cells. Nutr Res 2016; 36:731-41. [PMID: 27262535 DOI: 10.1016/j.nutres.2016.02.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 02/12/2016] [Accepted: 02/25/2016] [Indexed: 01/08/2023]
Abstract
In this study, we examined the cell differentiation effect of an ethanol extract of Cyclolepis genistoides D. Don, a herbaceous perennial belonging to the family Asteraceae (vernacular name: palo azul). Palo azul has numerous physiological effects that contribute to the prevention of metabolic syndromes, although the mechanism remains unclear. We previously suggested that palo azul has antidiabetic activity via an adipose differentiation effect. Here, we focused on whether palo azul promoted the differentiation of myoblasts. The mouse muscle myoblast cell line C2C12 was cultured and differentiated using horse serum with or without an ethanol extract of palo azul (12.5-200 μg/mL). Quantitative real-time polymerase chain reaction was performed to evaluate differentiation markers, including insulin-like growth factor-1 and myogenin. To evaluate myotube formation, myosin heavy-chain (MHC) expression and localization were detected by immunohistochemistry. Palo azul increased the expression of the differentiation markers. Furthermore, immunohistochemistry analysis revealed increased formation of MHC myotubes after palo azul treatment along with increased diameter and fusion indices of the myotubes. The expression level of MHC was also increased. In conclusion, palo azul may increase muscle mass in the body and improve insulin resistance conditions by facilitating the formation of myotubes by promoting myocyte differentiation.
Collapse
Affiliation(s)
- Hiromi Sato
- Department of Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuou-ku, Chiba-shi, Chiba 260-8675, Japan.
| | - Asami Funaki
- Department of Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuou-ku, Chiba-shi, Chiba 260-8675, Japan
| | - Yuki Kimura
- Department of Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuou-ku, Chiba-shi, Chiba 260-8675, Japan
| | - Mai Sumitomo
- Department of Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuou-ku, Chiba-shi, Chiba 260-8675, Japan
| | - Hiroya Yoshida
- IHM Inc, 7-22-17, Nishigotanda, Shinagawa-ku, Tokyo 141-0031, Japan
| | - Hideki Fukata
- JPD Co. Ltd, 7-98, Kitaitami, Itami-shi, Hyogo 664-0831, Japan
| | - Koichi Ueno
- Center of Preventive Medical Science, Chiba University, 1-8-1 Inohana, Chuou-ku, Chiba-shi, Chiba 260-8675, Japan
| |
Collapse
|
182
|
Ma Y, Hu J, Zhang N, Dong X, Li Y, Yang B, Tian W, Wang X. Prediction of Candidate Drugs for Treating Pancreatic Cancer by Using a Combined Approach. PLoS One 2016; 11:e0149896. [PMID: 26910401 PMCID: PMC4765895 DOI: 10.1371/journal.pone.0149896] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 02/05/2016] [Indexed: 01/15/2023] Open
Abstract
Pancreatic cancer is the leading cause of death from solid malignancies worldwide. Currently, gemcitabine is the only drug approved for treating pancreatic cancer. Developing new therapeutic drugs for this disease is, therefore, an urgent need. The C-Map project has provided a wealth of gene expression data that can be mined for repositioning drugs, a promising approach to new drug discovery. Typically, a drug is considered potentially useful for treating a disease if the drug-induced differential gene expression profile is negatively correlated with the differentially expressed genes in the target disease. However, many of the potentially useful drugs (PUDs) identified by gene expression profile correlation are likely false positives because, in C-Map, the cultured cell lines to which the drug is applied are not derived from diseased tissues. To solve this problem, we developed a combined approach for predicting candidate drugs for treating pancreatic cancer. We first identified PUDs for pancreatic cancer by using C-Map-based gene expression correlation analyses. We then applied an algorithm (Met-express) to predict key pancreatic cancer (KPC) enzymes involved in pancreatic cancer metabolism. Finally, we selected candidates from the PUDs by requiring that their targets be KPC enzymes or the substrates/products of KPC enzymes. Using this combined approach, we predicted seven candidate drugs for treating pancreatic cancer, three of which are supported by literature evidence, and three were experimentally validated to be inhibitory to pancreatic cancer celllines.
Collapse
Affiliation(s)
- Yanfen Ma
- Department of Clinical Laboratory, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi province, P.R. China
- Health Science Center of Xi'an Jiaotong University, Xi'an, Shaanxi province, P.R. China
| | - Jian Hu
- Department of Clinical Laboratory, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi province, P.R. China
| | - Ning Zhang
- Department of Clinical Laboratory, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi province, P.R. China
- Health Science Center of Xi'an Jiaotong University, Xi'an, Shaanxi province, P.R. China
| | - Xinran Dong
- Department of Biostatistics and Computational Biology, School of Life Science, Fudan University, Shanghai, China
| | - Ying Li
- Health Science Center of Xi'an Jiaotong University, Xi'an, Shaanxi province, P.R. China
- SHAANXI Kang Fu Hospital, Xi'an, Shaanxi province, P.R. China
| | - Bo Yang
- Department of Clinical Laboratory, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi province, P.R. China
| | - Weidong Tian
- Department of Biostatistics and Computational Biology, School of Life Science, Fudan University, Shanghai, China
| | - Xiaoqin Wang
- Department of Clinical Laboratory, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi province, P.R. China
| |
Collapse
|
183
|
Brook MS, Wilkinson DJ, Phillips BE, Perez-Schindler J, Philp A, Smith K, Atherton PJ. Skeletal muscle homeostasis and plasticity in youth and ageing: impact of nutrition and exercise. Acta Physiol (Oxf) 2016; 216:15-41. [PMID: 26010896 PMCID: PMC4843955 DOI: 10.1111/apha.12532] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Revised: 11/10/2014] [Accepted: 05/18/2015] [Indexed: 12/18/2022]
Abstract
Skeletal muscles comprise a substantial portion of whole body mass and are integral for locomotion and metabolic health. Increasing age is associated with declines in both muscle mass and function (e.g. strength‐related performance, power) with declines in muscle function quantitatively outweighing those in muscle volume. The mechanisms behind these declines are multi‐faceted involving both intrinsic age‐related metabolic dysregulation and environmental influences such as nutritional and physical activity. Ageing is associated with a degree of ‘anabolic resistance’ to these key environmental inputs, which likely accelerates the intrinsic processes driving ageing. On this basis, strategies to sensitize and/or promote anabolic responses to nutrition and physical activity are likely to be imperative in alleviating the progression and trajectory of sarcopenia. Both resistance‐ and aerobic‐type exercises are likely to confer functional and health benefits in older age, and a clutch of research suggests that enhancement of anabolic responsiveness to exercise and/or nutrition may be achieved by optimizing modifications of muscle‐loading paradigms (workload, volume, blood flow restriction) or nutritional support (e.g. essential amino acid/leucine) patterns. Nonetheless, more work is needed in which a more holistic view in ageing studies is taken into account. This should include improved characterization of older study recruits, that is physical activity/nutritional behaviours, to limit confounding variables influencing whether findings are attributable to age, or other environmental influences. Nonetheless, on balance, ageing is associated with declines in muscle mass and function and a partially related decline in aerobic capacity. There is also good evidence that metabolic flexibility is impaired in older age.
Collapse
Affiliation(s)
- M. S. Brook
- MRC-ARUK Centre of Excellence for Musculoskeletal Ageing Research, Clinical Metabolic and Molecular Physiology; University of Nottingham; Royal Derby Hospital Centre; Derby UK
| | - D. J. Wilkinson
- MRC-ARUK Centre of Excellence for Musculoskeletal Ageing Research, Clinical Metabolic and Molecular Physiology; University of Nottingham; Royal Derby Hospital Centre; Derby UK
| | - B. E. Phillips
- MRC-ARUK Centre of Excellence for Musculoskeletal Ageing Research, Clinical Metabolic and Molecular Physiology; University of Nottingham; Royal Derby Hospital Centre; Derby UK
| | - J. Perez-Schindler
- MRC-ARUK Centre of Excellence for Musculoskeletal Ageing Research, School of Sport, Exercise and Rehabilitation Sciences; University of Birmingham; Birmingham UK
| | - A. Philp
- MRC-ARUK Centre of Excellence for Musculoskeletal Ageing Research, School of Sport, Exercise and Rehabilitation Sciences; University of Birmingham; Birmingham UK
| | - K. Smith
- MRC-ARUK Centre of Excellence for Musculoskeletal Ageing Research, Clinical Metabolic and Molecular Physiology; University of Nottingham; Royal Derby Hospital Centre; Derby UK
| | - P. J. Atherton
- MRC-ARUK Centre of Excellence for Musculoskeletal Ageing Research, Clinical Metabolic and Molecular Physiology; University of Nottingham; Royal Derby Hospital Centre; Derby UK
| |
Collapse
|
184
|
Caloric restriction and exercise "mimetics'': Ready for prime time? Pharmacol Res 2015; 103:158-66. [PMID: 26658171 DOI: 10.1016/j.phrs.2015.11.009] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Accepted: 11/19/2015] [Indexed: 12/19/2022]
Abstract
Exercise and diet are powerful interventions to prevent and ameliorate various pathologies. The development of pharmacological agents that confer exercise- or caloric restriction-like phenotypic effects is thus an appealing therapeutic strategy in diseases or even when used as life-style and longevity drugs. Such so-called exercise or caloric restriction "mimetics" have so far mostly been described in pre-clinical, experimental settings with limited translation into humans. Interestingly, many of these compounds activate related signaling pathways, most often postulated to act on the common downstream effector peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α) in skeletal muscle. In this review, resveratrol and other exercise- and caloric restriction "mimetics" are discussed with a special focus on feasibility, chances and limitations of using such compounds in patients as well as in healthy individuals.
Collapse
|
185
|
Jeong JW, Shim JJ, Choi ID, Kim SH, Ra J, Ku HK, Lee DE, Kim TY, Jeung W, Lee JH, Lee KW, Huh CS, Sim JH, Ahn YT. Apple Pomace Extract Improves Endurance in Exercise Performance by Increasing Strength and Weight of Skeletal Muscle. J Med Food 2015; 18:1380-6. [PMID: 26331671 DOI: 10.1089/jmf.2014.3401] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Ursolic acid is a lipophilic pentacyclic triterpenoid found in many fruits and herbs and is used in several herbal folk medicines for diabetes. In this study, we evaluated the effects of apple pomace extract (APE; ursolic acid content, 183 mg/g) on skeletal muscle atrophy. To examine APE therapeutic potential in muscle atrophy, we investigated APE effects on the expression of biomarkers associated with muscle atrophy and hypertrophy. We found that APE inhibited atrophy, while inducing hypertrophy in C2C12 myotubes by decreasing the expression of atrophy-related genes and increasing the expression of hypertrophy-associated genes. The in vivo experiments using mice fed a diet with or without APE showed that APE intake increased skeletal muscle mass, as well as grip strength and exercise capacity. In addition, APE significantly improved endurance in the mice, as evidenced by increased exhaustive running time and muscle weight, and reduced the expression of the genes involved in the development of muscle atrophy. APE also decreased the concentration of serum lactate and lactate dehydrogenase, inorganic phosphate, and creatinine, the indicators of accumulated fatigue and exercise-induced stress. These results suggest that APE may be useful as an ergogenic functional food or dietary supplement.
Collapse
Affiliation(s)
- Ji-Woong Jeong
- 1 R&BD Center, Korea Yakult Co, Ltd. , Yongin, Republic of Korea
| | - Jae-Jung Shim
- 1 R&BD Center, Korea Yakult Co, Ltd. , Yongin, Republic of Korea
- 2 College of Agriculture and Life Sciences, Seoul National University , Seoul, Republic of Korea
| | - Il-Dong Choi
- 1 R&BD Center, Korea Yakult Co, Ltd. , Yongin, Republic of Korea
| | - Sung-Hwan Kim
- 1 R&BD Center, Korea Yakult Co, Ltd. , Yongin, Republic of Korea
| | - Jehyeon Ra
- 1 R&BD Center, Korea Yakult Co, Ltd. , Yongin, Republic of Korea
| | - Hyung Keun Ku
- 1 R&BD Center, Korea Yakult Co, Ltd. , Yongin, Republic of Korea
| | - Dong Eun Lee
- 1 R&BD Center, Korea Yakult Co, Ltd. , Yongin, Republic of Korea
| | - Tae-Youl Kim
- 1 R&BD Center, Korea Yakult Co, Ltd. , Yongin, Republic of Korea
| | - Woonhee Jeung
- 1 R&BD Center, Korea Yakult Co, Ltd. , Yongin, Republic of Korea
| | - Jung-Hee Lee
- 1 R&BD Center, Korea Yakult Co, Ltd. , Yongin, Republic of Korea
| | - Ki Won Lee
- 2 College of Agriculture and Life Sciences, Seoul National University , Seoul, Republic of Korea
| | - Chul-Sung Huh
- 3 Graduate School of International Agricultural Technology, Institute of Green Bio Science and Technology, Seoul National University , Pyeongchang, Republic of Korea
| | - Jae-Hun Sim
- 1 R&BD Center, Korea Yakult Co, Ltd. , Yongin, Republic of Korea
| | - Young-Tae Ahn
- 1 R&BD Center, Korea Yakult Co, Ltd. , Yongin, Republic of Korea
| |
Collapse
|
186
|
Woźniak Ł, Skąpska S, Marszałek K. Ursolic Acid--A Pentacyclic Triterpenoid with a Wide Spectrum of Pharmacological Activities. Molecules 2015; 20:20614-41. [PMID: 26610440 PMCID: PMC6332387 DOI: 10.3390/molecules201119721] [Citation(s) in RCA: 228] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Revised: 08/21/2015] [Accepted: 09/23/2015] [Indexed: 12/12/2022] Open
Abstract
Ursolic acid (UA) is a natural terpene compound exhibiting many pharmaceutical properties. In this review the current state of knowledge about the health-promoting properties of this widespread, biologically active compound, as well as information about its occurrence and biosynthesis are presented. Particular attention has been paid to the application of ursolic acid as an anti-cancer agent; it is worth noticing that clinical tests suggesting the possibility of practical use of UA have already been conducted. Amongst other pharmacological properties of UA one can mention protective effect on lungs, kidneys, liver and brain, anti-inflammatory properties, anabolic effects on skeletal muscles and the ability to suppress bone density loss leading to osteoporosis. Ursolic acid also exhibits anti-microbial features against numerous strains of bacteria, HIV and HCV viruses and Plasmodium protozoa causing malaria.
Collapse
Affiliation(s)
- Łukasz Woźniak
- Department of Fruit and Vegetable Product Technology, Institute of Agricultural and Food Biotechnology, 36 Rakowiecka Street, 02-532 Warsaw, Poland.
| | - Sylwia Skąpska
- Department of Fruit and Vegetable Product Technology, Institute of Agricultural and Food Biotechnology, 36 Rakowiecka Street, 02-532 Warsaw, Poland.
| | - Krystian Marszałek
- Department of Fruit and Vegetable Product Technology, Institute of Agricultural and Food Biotechnology, 36 Rakowiecka Street, 02-532 Warsaw, Poland.
| |
Collapse
|
187
|
Ebert SM, Dyle MC, Bullard SA, Dierdorff JM, Murry DJ, Fox DK, Bongers KS, Lira VA, Meyerholz DK, Talley JJ, Adams CM. Identification and Small Molecule Inhibition of an Activating Transcription Factor 4 (ATF4)-dependent Pathway to Age-related Skeletal Muscle Weakness and Atrophy. J Biol Chem 2015; 290:25497-511. [PMID: 26338703 PMCID: PMC4646196 DOI: 10.1074/jbc.m115.681445] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Revised: 08/30/2015] [Indexed: 12/17/2022] Open
Abstract
Aging reduces skeletal muscle mass and strength, but the underlying molecular mechanisms remain elusive. Here, we used mouse models to investigate molecular mechanisms of age-related skeletal muscle weakness and atrophy as well as new potential interventions for these conditions. We identified two small molecules that significantly reduce age-related deficits in skeletal muscle strength, quality, and mass: ursolic acid (a pentacyclic triterpenoid found in apples) and tomatidine (a steroidal alkaloid derived from green tomatoes). Because small molecule inhibitors can sometimes provide mechanistic insight into disease processes, we used ursolic acid and tomatidine to investigate the pathogenesis of age-related muscle weakness and atrophy. We found that ursolic acid and tomatidine generate hundreds of small positive and negative changes in mRNA levels in aged skeletal muscle, and the mRNA expression signatures of the two compounds are remarkably similar. Interestingly, a subset of the mRNAs repressed by ursolic acid and tomatidine in aged muscle are positively regulated by activating transcription factor 4 (ATF4). Based on this finding, we investigated ATF4 as a potential mediator of age-related muscle weakness and atrophy. We found that a targeted reduction in skeletal muscle ATF4 expression reduces age-related deficits in skeletal muscle strength, quality, and mass, similar to ursolic acid and tomatidine. These results elucidate ATF4 as a critical mediator of age-related muscle weakness and atrophy. In addition, these results identify ursolic acid and tomatidine as potential agents and/or lead compounds for reducing ATF4 activity, weakness, and atrophy in aged skeletal muscle.
Collapse
Affiliation(s)
- Scott M Ebert
- From the Departments of Internal Medicine, the Fraternal Order of Eagles Diabetes Research Center, and the Iowa City Veterans Affairs Medical Center, Iowa City, Iowa 52246, and Emmyon, Inc., Coralville, Iowa 52241
| | - Michael C Dyle
- From the Departments of Internal Medicine, the Fraternal Order of Eagles Diabetes Research Center, and the Iowa City Veterans Affairs Medical Center, Iowa City, Iowa 52246, and Molecular Physiology and Biophysics
| | - Steven A Bullard
- From the Departments of Internal Medicine, the Fraternal Order of Eagles Diabetes Research Center, and the Iowa City Veterans Affairs Medical Center, Iowa City, Iowa 52246, and
| | - Jason M Dierdorff
- From the Departments of Internal Medicine, the Fraternal Order of Eagles Diabetes Research Center, and the Iowa City Veterans Affairs Medical Center, Iowa City, Iowa 52246, and
| | - Daryl J Murry
- the College of Pharmacy, The University of Iowa, Iowa City, Iowa 52242
| | - Daniel K Fox
- From the Departments of Internal Medicine, the Fraternal Order of Eagles Diabetes Research Center, and the Iowa City Veterans Affairs Medical Center, Iowa City, Iowa 52246, and Molecular Physiology and Biophysics
| | - Kale S Bongers
- From the Departments of Internal Medicine, the Fraternal Order of Eagles Diabetes Research Center, and the Iowa City Veterans Affairs Medical Center, Iowa City, Iowa 52246, and Molecular Physiology and Biophysics
| | - Vitor A Lira
- the Fraternal Order of Eagles Diabetes Research Center, and Health and Human Physiology, and
| | | | | | - Christopher M Adams
- From the Departments of Internal Medicine, the Fraternal Order of Eagles Diabetes Research Center, and the Iowa City Veterans Affairs Medical Center, Iowa City, Iowa 52246, and Emmyon, Inc., Coralville, Iowa 52241 Molecular Physiology and Biophysics,
| |
Collapse
|
188
|
Kibble M, Saarinen N, Tang J, Wennerberg K, Mäkelä S, Aittokallio T. Network pharmacology applications to map the unexplored target space and therapeutic potential of natural products. Nat Prod Rep 2015; 32:1249-66. [PMID: 26030402 DOI: 10.1039/c5np00005j] [Citation(s) in RCA: 288] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
It is widely accepted that drug discovery often requires a systems-level polypharmacology approach to tackle problems such as lack of efficacy and emerging resistance of single-targeted compounds. Network pharmacology approaches are increasingly being developed and applied to find new therapeutic opportunities and to re-purpose approved drugs. However, these recent advances have been relatively slow to be translated into the field of natural products. Here, we argue that a network pharmacology approach would enable an effective mapping of the yet unexplored target space of natural products, hence providing a systematic means to extend the druggable space of proteins implicated in various complex diseases. We give an overview of the key network pharmacology concepts and recent experimental-computational approaches that have been successfully applied to natural product research, including unbiased elucidation of mechanisms of action as well as systematic prediction of effective therapeutic combinations. We focus specifically on anticancer applications that use in vivo and in vitro functional phenotypic measurements, such as genome-wide transcriptomic response profiles, which enable a global modelling of the multi-target activity at the level of the biological pathways and interaction networks. We also provide representative examples of other disease applications, databases and tools as well as existing and emerging resources, which may prove useful for future natural product research. Finally, we offer our personal view of the current limitations, prospective developments and open questions in this exciting field.
Collapse
Affiliation(s)
- Milla Kibble
- Institute for Molecular Medicine Finland (FIMM), Biomedicum Helsinki 2U, 00014 University of Helsinki, Finland.
| | | | | | | | | | | |
Collapse
|
189
|
Iorio F, Shrestha RL, Levin N, Boilot V, Garnett MJ, Saez-Rodriguez J, Draviam VM. A Semi-Supervised Approach for Refining Transcriptional Signatures of Drug Response and Repositioning Predictions. PLoS One 2015; 10:e0139446. [PMID: 26452147 PMCID: PMC4599732 DOI: 10.1371/journal.pone.0139446] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 08/11/2015] [Indexed: 11/28/2022] Open
Abstract
We present a novel strategy to identify drug-repositioning opportunities. The starting point of our method is the generation of a signature summarising the consensual transcriptional response of multiple human cell lines to a compound of interest (namely the seed compound). This signature can be derived from data in existing databases, such as the connectivity-map, and it is used at first instance to query a network interlinking all the connectivity-map compounds, based on the similarity of their transcriptional responses. This provides a drug neighbourhood, composed of compounds predicted to share some effects with the seed one. The original signature is then refined by systematically reducing its overlap with the transcriptional responses induced by drugs in this neighbourhood that are known to share a secondary effect with the seed compound. Finally, the drug network is queried again with the resulting refined signatures and the whole process is carried on for a number of iterations. Drugs in the final refined neighbourhood are then predicted to exert the principal mode of action of the seed compound. We illustrate our approach using paclitaxel (a microtubule stabilising agent) as seed compound. Our method predicts that glipizide and splitomicin perturb microtubule function in human cells: a result that could not be obtained through standard signature matching methods. In agreement, we find that glipizide and splitomicin reduce interphase microtubule growth rates and transiently increase the percentage of mitotic cells-consistent with our prediction. Finally, we validated the refined signatures of paclitaxel response by mining a large drug screening dataset, showing that human cancer cell lines whose basal transcriptional profile is anti-correlated to them are significantly more sensitive to paclitaxel and docetaxel.
Collapse
Affiliation(s)
- Francesco Iorio
- European Molecular Biology Laboratory–European Bioinformatics institute, Wellcome Trust Genome Campus, CB10 1SD, Cambridge, United Kingdom
| | - Roshan L. Shrestha
- Department of Genetics—University of Cambridge, Downing Street, CB2 3EH, Cambridge, United Kingdom
| | - Nicolas Levin
- Department of Genetics—University of Cambridge, Downing Street, CB2 3EH, Cambridge, United Kingdom
| | - Viviane Boilot
- Department of Genetics—University of Cambridge, Downing Street, CB2 3EH, Cambridge, United Kingdom
| | - Mathew J. Garnett
- Cancer genome project–Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, CB10 1SD, Cambridge, United Kingdom
| | - Julio Saez-Rodriguez
- European Molecular Biology Laboratory–European Bioinformatics institute, Wellcome Trust Genome Campus, CB10 1SD, Cambridge, United Kingdom
- RWTH-Aachen University Hospital, Templergraben 55, 52062, Aachen, Germany
| | - Viji M. Draviam
- Department of Genetics—University of Cambridge, Downing Street, CB2 3EH, Cambridge, United Kingdom
| |
Collapse
|
190
|
Smalley JL, Breda C, Mason RP, Kooner G, Luthi-Carter R, Gant TW, Giorgini F. Connectivity mapping uncovers small molecules that modulate neurodegeneration in Huntington's disease models. J Mol Med (Berl) 2015; 94:235-45. [PMID: 26428929 PMCID: PMC4762922 DOI: 10.1007/s00109-015-1344-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 08/24/2015] [Accepted: 09/09/2015] [Indexed: 12/18/2022]
Abstract
UNLABELLED Huntington's disease (HD) is a genetic disease caused by a CAG trinucleotide repeat expansion encoding a polyglutamine tract in the huntingtin (HTT) protein, ultimately leading to neuronal loss and consequent cognitive decline and death. As no treatments for HD currently exist, several chemical screens have been performed using cell-based models of mutant HTT toxicity. These screens measured single disease-related endpoints, such as cell death, but had low 'hit rates' and limited dimensionality for therapeutic detection. Here, we have employed gene expression microarray analysis of HD samples--a snapshot of the expression of 25,000 genes--to define a gene expression signature for HD from publically available data. We used this information to mine a database for chemicals positively and negatively correlated to the HD gene expression signature using the Connectivity Map, a tool for comparing large sets of gene expression patterns. Chemicals with negatively correlated expression profiles were highly enriched for protective characteristics against mutant HTT fragment toxicity in in vitro and in vivo models. This study demonstrates the potential of using gene expression to mine chemical activity, guide chemical screening, and detect potential novel therapeutic compounds. KEY MESSAGES Single-endpoint chemical screens have low therapeutic discovery hit-rates. In the context of HD, we guided a chemical screen using gene expression data. The resulting chemicals were highly enriched for suppressors of mutant HTT fragment toxicity. This study provides a proof of concept for wider usage in all chemical screening.
Collapse
Affiliation(s)
- Joshua L Smalley
- Department of Genetics, University of Leicester, Leicester, LE1 7RH, UK.,MRC Toxicology Unit, University of Leicester, Leicester, LE1 7HB, UK
| | - Carlo Breda
- Department of Genetics, University of Leicester, Leicester, LE1 7RH, UK
| | - Robert P Mason
- Department of Genetics, University of Leicester, Leicester, LE1 7RH, UK
| | - Gurdeep Kooner
- Department of Genetics, University of Leicester, Leicester, LE1 7RH, UK
| | - Ruth Luthi-Carter
- Department of Cell Physiology and Pharmacology, University of Leicester, Leicester, LE1 7RH, UK
| | - Timothy W Gant
- MRC Toxicology Unit, University of Leicester, Leicester, LE1 7HB, UK.,Centre for Radiation, Chemical and Environmental Hazards, Public Health England, Harwell Campus, Oxfordshire, OX11 0RQ, UK
| | - Flaviano Giorgini
- Department of Genetics, University of Leicester, Leicester, LE1 7RH, UK.
| |
Collapse
|
191
|
The use of gene arrays and corresponding connectivity mapping (Cmap) to identify novel anti-ageing ingredients. Int J Cosmet Sci 2015; 37 Suppl 1:9-14. [DOI: 10.1111/ics.12251] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 06/05/2015] [Indexed: 01/10/2023]
|
192
|
Suneja M, Fox DK, Fink BD, Herlein JA, Adams CM, Sivitz WI. Evidence for metabolic aberrations in asymptomatic persons with type 2 diabetes after initiation of simvastatin therapy. Transl Res 2015; 166:176-87. [PMID: 25683525 PMCID: PMC4509977 DOI: 10.1016/j.trsl.2015.01.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Revised: 01/03/2015] [Accepted: 01/20/2015] [Indexed: 01/14/2023]
Abstract
Hydroxymethylglutaryl coenzyme A reductase inhibitors (statins) prevent vascular events and are widely prescribed, particularly in persons with type 2 diabetes. However, intolerability because of myopathic symptoms often limits their use. We investigated the effects of simvastatin on parameters of mitochondrial function and muscle gene expression in 11 subjects with type 2 diabetes, none of whom had statin intolerance. After withdrawal of statins for 2 months, we obtained blood samples, performed vastus lateralis muscle biopsies, and assessed whole body resting energy expenditure (REE). We then reinitiated therapy using simvastatin, 20 mg/d, for 1 month before repeating these studies. As expected, simvastatin lowered low-density lipoprotein, but did not induce myalgias or significant increases in serum creatine kinase. However, we found subtle but significant reductions in muscle citrate synthase activity and REE. In addition, quantitative polymerase chain reaction and gene set enrichment analysis of muscle samples revealed significantly repressed gene sets involved in mitochondrial function and induced gene sets involved in remodeling of the extracellular matrix. Furthermore, the effects of simvastatin on muscle gene sets showed some similarities to previously described changes that occur in Duchenne muscular dystrophy, polymyositis, and dermatomyositis. Although statins inhibit an early step in coenzyme Q (CoQ) biosynthesis, we observed no differences in CoQ content within skeletal muscle mitochondria, muscle tissue, or circulating platelets. In summary, we report subtle changes in whole body energetics, mitochondrial citrate synthase activity, and microarray data consistent with subclinical myopathy. Although the benefits of statin therapy are clear, further understanding of muscular perturbations should help guide safety and tolerability.
Collapse
Affiliation(s)
- Manish Suneja
- Division of Nephrology, Department of Internal Medicine, University of Iowa and the Iowa City Veterans Affairs Health Care System, Iowa City VA, Iowa City, Iowa
| | - Daniel K Fox
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, Iowa
| | - Brian D Fink
- Division of Endocrinology and Metabolism, Department of Internal Medicine, University of Iowa and the Iowa City Veterans Affairs Health Care System, Iowa City VA, Iowa City, Iowa
| | - Judy A Herlein
- Division of Endocrinology and Metabolism, Department of Internal Medicine, University of Iowa and the Iowa City Veterans Affairs Health Care System, Iowa City VA, Iowa City, Iowa
| | - Christopher M Adams
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, Iowa; Division of Endocrinology and Metabolism, Department of Internal Medicine, University of Iowa and the Iowa City Veterans Affairs Health Care System, Iowa City VA, Iowa City, Iowa
| | - William I Sivitz
- Division of Endocrinology and Metabolism, Department of Internal Medicine, University of Iowa and the Iowa City Veterans Affairs Health Care System, Iowa City VA, Iowa City, Iowa.
| |
Collapse
|
193
|
Sung B, Hwang SY, Kim MJ, Kim M, Jeong JW, Kim CM, Chung HY, Kim ND. Loquat leaf extract enhances myogenic differentiation, improves muscle function and attenuates muscle loss in aged rats. Int J Mol Med 2015; 36:792-800. [PMID: 26178971 DOI: 10.3892/ijmm.2015.2286] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2015] [Accepted: 07/03/2015] [Indexed: 11/06/2022] Open
Abstract
A main characteristic of aging is the debilitating, progressive and generalized impairment of biological functions, resulting in an increased vulnerability to disease and death. Skeletal muscle comprises approximately 40% of the human body; thus, it is the most abundant tissue. At the age of 30 onwards, 0.5‑1% of human muscle mass is lost each year, with a marked acceleration in the rate of decline after the age of 65. Thus, novel strategies that effectively attenuate skeletal muscle loss and enhance muscle function are required to improve the quality of life of older subjects. The aim of the present study was to determine whether loquat (Eriobotrya japonica) leaf extract (LE) can prevent the loss of skeletal muscle function in aged rats. Young (5-month-old) and aged (18‑19-month-old) rats were fed LE (50 mg/kg/day) for 35 days and the changes in muscle mass and strength were evaluated. The age‑associated loss of grip strength was attenuated, and muscle mass and muscle creatine kinase (CK) activity were enhanced following the administration of LE. Histochemical analysis also revealed that LE abrogated the age‑associated decrease in cross‑sectional area (CSA) and decreased the amount of connective tissue in the muscle of aged rats. To investigate the mode of action of LE, C2C12 murine myoblasts were used to evaluate the myogenic potential of LE. The expression levels of myogenic proteins (MyoD and myogenin) and functional myosin heavy chain (MyHC) were measured by western blot analysis. LE enhanced MyoD, myogenin and MyHC expression. The changes in the expression of myogenic genes corresponded with an increase in the activity of CK, a myogenic differentiation marker. Finally, LE activated the Akt/mammalian target of rapamycin (mTOR) signaling pathway, which is involved in muscle protein synthesis during myogenesis. These findings suggest that LE attenuates sarcopenia by promoting myogenic differentiation and subsequently promoting muscle protein synthesis.
Collapse
Affiliation(s)
- Bokyung Sung
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan 609‑735, Republic of Korea
| | - Seong Yeon Hwang
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan 609‑735, Republic of Korea
| | - Min Jo Kim
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan 609‑735, Republic of Korea
| | - Minjung Kim
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan 609‑735, Republic of Korea
| | - Ji Won Jeong
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan 609‑735, Republic of Korea
| | - Cheol Min Kim
- Research Center for Anti‑Aging Technology Development, Pusan National University, Busan 609‑735, Republic of Korea
| | - Hae Young Chung
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan 609‑735, Republic of Korea
| | - Nam Deuk Kim
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan 609‑735, Republic of Korea
| |
Collapse
|
194
|
Bakhtiari N, Hosseinkhani S, Tashakor A, Hemmati R. Ursolic acid ameliorates aging-metabolic phenotype through promoting of skeletal muscle rejuvenation. Med Hypotheses 2015; 85:1-6. [PMID: 25976755 DOI: 10.1016/j.mehy.2015.02.014] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2014] [Revised: 02/17/2015] [Accepted: 02/25/2015] [Indexed: 12/25/2022]
Abstract
Ursolic acid (UA) is a lipophilic compound, which highly found in apple peels. UA has some certain features, of the most important is its anabolic effects on skeletal muscles, which in turn plays a prominent role in the aging process, encouraged us to evaluate skeletal muscle rejuvenation. This study seeks to address the two following questions: primarily, we wonder to know if UA increases anti-aging biomarkers (SIRT1 and PGC-1α) in the isolated satellite cells, to pave the way for satellite cells proliferation. The results revealed that UA elevated the expression of SIRT1 (∼ 35 folds) and PGC-1α (∼ 175 folds) genes. The other question that needs to be asked, however, is to understand whether it is possible to generalize the in vitro findings to in vivo. For this, a study was designed to investigate the effects of UA on the cellular energy status in the animal models (C57BL/6 mice). We found that UA decreased cellular energy charges such as ATP (∼ 3 times) and ADP (∼ 18 times). With respect to the role of UA in energy expenditure and as an anti-aging biomarker, one might wonder to elucidate skeletal muscle rejuvenation as well as satellite cells proliferation and neomyogenesis. The results illustrated that UA boosted neomyogenesis through enhancing the number of satellite cells. In addition, rejuvenation effects of UA on the skeletal muscle promptly encouraged us to reexamine the performance of skeletal muscles. The results indicated that UA through increasing myoglobin expression (∼ 2 folds) accompanied with transforming of glycolytic to fast oxidative status chiefly and slow-twitch muscle fibers. To the best of our knowledge, it seems that UA might be considered as a potential candidate for treatment of pathological conditions associated with muscular atrophy and dysfunction, including skeletal muscle atrophy, amyotrophic lateral sclerosis (ALS), sarcopenia and metabolic diseases of the muscles.
Collapse
Affiliation(s)
- Nuredin Bakhtiari
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Saman Hosseinkhani
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Amin Tashakor
- Department of Biochemistry, Institute of Biochemistry and Biophysics, University of Tehran, Iran
| | - Roohullah Hemmati
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
195
|
Manzotti G, Parenti S, Ferrari-Amorotti G, Soliera AR, Cattelani S, Montanari M, Cavalli D, Ertel A, Grande A, Calabretta B. Monocyte-macrophage differentiation of acute myeloid leukemia cell lines by small molecules identified through interrogation of the Connectivity Map database. Cell Cycle 2015; 14:2578-89. [PMID: 26102293 DOI: 10.1080/15384101.2015.1033591] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
The transcription factor C/EBPα is required for granulocytic differentiation of normal myeloid progenitors and is frequently inactivated in acute myeloid leukemia (AML) cells. Ectopic expression of C/EBPα in AML cells suppresses proliferation and induces differentiation suggesting that restoring C/EBPα expression/activity in AML cells could be therapeutically useful. Unfortunately, current approaches of gene or protein delivery in leukemic cells are unsatisfactory. However, "drug repurposing" is becoming a very attractive strategy to identify potential new uses for existing drugs. In this study, we assessed the biological effects of candidate C/EBPα-mimetics identified by interrogation of the Connectivity Map database. We found that amantadine, an antiviral and anti-Parkinson agent, induced a monocyte-macrophage-like differentiation of HL60, U937, Kasumi-1 myeloid leukemia cell lines, as indicated by morphology and differentiation antigen expression, when used in combination with suboptimal concentration of all trans retinoic acid (ATRA) or Vit D3. The effect of amantadine depends, in part, on increased activity of the vitamin D receptor (VDR), since it induced VDR expression and amantadine-dependent monocyte-macrophage differentiation of HL60 cells was blocked by expression of dominant-negative VDR. These results reveal a new function for amantadine and support the concept that screening of the Connectivity Map database can identify small molecules that mimic the effect of transcription factors required for myelo-monocytic differentiation.
Collapse
Affiliation(s)
- Gloria Manzotti
- a Department of Diagnostic and Clinical Medicine and Public Health ; University of Modena and R. Emilia ; Modena , Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
196
|
NOH KYUNGKYUN, CHUNG KIWUNG, SUNG BOKYUNG, KIM MINJO, PARK CHANHUM, YOON CHANGSHIN, CHOI JAESUE, KIM MIKYUNG, KIM CHEOLMIN, KIM NAMDEUK, CHUNG HAEYOUNG. Loquat (Eriobotrya japonica) extract prevents dexamethasone-induced muscle atrophy by inhibiting the muscle degradation pathway in Sprague Dawley rats. Mol Med Rep 2015; 12:3607-3614. [DOI: 10.3892/mmr.2015.3821] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2014] [Accepted: 05/08/2015] [Indexed: 11/06/2022] Open
|
197
|
Barreiro E, Sznajder JI, Nader GA, Budinger GRS. Muscle dysfunction in patients with lung diseases: a growing epidemic. Am J Respir Crit Care Med 2015; 191:616-9. [PMID: 25767924 DOI: 10.1164/rccm.201412-2189oe] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Affiliation(s)
- Esther Barreiro
- 1 Respiratory Medicine Department-Lung Cancer Research Group, Institute of Medical Research of Hospital del Mar (IMIM)-Hospital del Mar, Parc de Salut Mar, Barcelona Biomedical Research Park, Barcelona, Spain
| | | | | | | |
Collapse
|
198
|
Robriquet F, Lardenois A, Babarit C, Larcher T, Dubreil L, Leroux I, Zuber C, Ledevin M, Deschamps JY, Fromes Y, Cherel Y, Guevel L, Rouger K. Differential Gene Expression Profiling of Dystrophic Dog Muscle after MuStem Cell Transplantation. PLoS One 2015; 10:e0123336. [PMID: 25955839 PMCID: PMC4425432 DOI: 10.1371/journal.pone.0123336] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 03/02/2015] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Several adult stem cell populations exhibit myogenic regenerative potential, thus representing attractive candidates for therapeutic approaches of neuromuscular diseases such as Duchenne Muscular Dystrophy (DMD). We have recently shown that systemic delivery of MuStem cells, skeletal muscle-resident stem cells isolated in healthy dog, generates the remodelling of muscle tissue and gives rise to striking clinical benefits in Golden Retriever Muscular Dystrophy (GRMD) dog. This global effect, which is observed in the clinically relevant DMD animal model, leads us to question here the molecular pathways that are impacted by MuStem cell transplantation. To address this issue, we compare the global gene expression profile between healthy, GRMD and MuStem cell treated GRMD dog muscle, four months after allogenic MuStem cell transplantation. RESULTS In the dystrophic context of the GRMD dog, disease-related deregulation is observed in the case of 282 genes related to various processes such as inflammatory response, regeneration, calcium ion binding, extracellular matrix organization, metabolism and apoptosis regulation. Importantly, we reveal the impact of MuStem cell transplantation on several molecular and cellular pathways based on a selection of 31 genes displaying signals specifically modulated by the treatment. Concomitant with a diffuse dystrophin expression, a histological remodelling and a stabilization of GRMD dog clinical status, we show that cell delivery is associated with an up-regulation of genes reflecting a sustained enhancement of muscle regeneration. We also identify a decreased mRNA expression of a set of genes having metabolic functions associated with lipid homeostasis and energy. Interestingly, ubiquitin-mediated protein degradation is highly enhanced in GRMD dog muscle after systemic delivery of MuStem cells. CONCLUSIONS Overall, our results provide the first high-throughput characterization of GRMD dog muscle and throw new light on the complex molecular/cellular effects associated with muscle repair and the clinical efficacy of MuStem cell-based therapy.
Collapse
Affiliation(s)
- Florence Robriquet
- INRA, UMR703 PAnTher, Nantes, France
- LUNAM Université, Oniris, École nationale vétérinaire, agro-alimentaire et de l’alimentation Nantes-Atlantique, Nantes, France
- Université de Nantes, Nantes, France
| | - Aurélie Lardenois
- INRA, UMR703 PAnTher, Nantes, France
- LUNAM Université, Oniris, École nationale vétérinaire, agro-alimentaire et de l’alimentation Nantes-Atlantique, Nantes, France
| | - Candice Babarit
- INRA, UMR703 PAnTher, Nantes, France
- LUNAM Université, Oniris, École nationale vétérinaire, agro-alimentaire et de l’alimentation Nantes-Atlantique, Nantes, France
| | - Thibaut Larcher
- INRA, UMR703 PAnTher, Nantes, France
- LUNAM Université, Oniris, École nationale vétérinaire, agro-alimentaire et de l’alimentation Nantes-Atlantique, Nantes, France
| | - Laurence Dubreil
- INRA, UMR703 PAnTher, Nantes, France
- LUNAM Université, Oniris, École nationale vétérinaire, agro-alimentaire et de l’alimentation Nantes-Atlantique, Nantes, France
| | - Isabelle Leroux
- INRA, UMR703 PAnTher, Nantes, France
- LUNAM Université, Oniris, École nationale vétérinaire, agro-alimentaire et de l’alimentation Nantes-Atlantique, Nantes, France
| | - Céline Zuber
- INRA, UMR703 PAnTher, Nantes, France
- LUNAM Université, Oniris, École nationale vétérinaire, agro-alimentaire et de l’alimentation Nantes-Atlantique, Nantes, France
| | - Mireille Ledevin
- INRA, UMR703 PAnTher, Nantes, France
- LUNAM Université, Oniris, École nationale vétérinaire, agro-alimentaire et de l’alimentation Nantes-Atlantique, Nantes, France
| | - Jack-Yves Deschamps
- INRA, UMR703 PAnTher, Nantes, France
- LUNAM Université, Oniris, École nationale vétérinaire, agro-alimentaire et de l’alimentation Nantes-Atlantique, Nantes, France
| | - Yves Fromes
- LUNAM Université, Oniris, École nationale vétérinaire, agro-alimentaire et de l’alimentation Nantes-Atlantique, Nantes, France
- Laboratoire RMN AIM-CEA, Institut de Myologie, Hôpital Pitié-Salpêtrière, Paris, France
| | - Yan Cherel
- INRA, UMR703 PAnTher, Nantes, France
- LUNAM Université, Oniris, École nationale vétérinaire, agro-alimentaire et de l’alimentation Nantes-Atlantique, Nantes, France
| | - Laetitia Guevel
- INRA, UMR703 PAnTher, Nantes, France
- LUNAM Université, Oniris, École nationale vétérinaire, agro-alimentaire et de l’alimentation Nantes-Atlantique, Nantes, France
- Université de Nantes, Nantes, France
- * E-mail:
| | - Karl Rouger
- INRA, UMR703 PAnTher, Nantes, France
- LUNAM Université, Oniris, École nationale vétérinaire, agro-alimentaire et de l’alimentation Nantes-Atlantique, Nantes, France
| |
Collapse
|
199
|
Adams CM, Ebert SM, Dyle MC. Use of mRNA expression signatures to discover small molecule inhibitors of skeletal muscle atrophy. Curr Opin Clin Nutr Metab Care 2015; 18:263-8. [PMID: 25807353 PMCID: PMC5512448 DOI: 10.1097/mco.0000000000000159] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PURPOSE OF REVIEW Here, we discuss a recently developed experimental strategy for discovering small molecules with potential to prevent and treat skeletal muscle atrophy. RECENT FINDINGS Muscle atrophy involves and requires widespread changes in skeletal muscle gene expression, which generate complex but measurable patterns of positive and negative changes in skeletal muscle mRNA levels (a.k.a. mRNA expression signatures of muscle atrophy). Many bioactive small molecules generate their own characteristic mRNA expression signatures, and by identifying small molecules whose signatures approximate mirror images of muscle atrophy signatures, one may identify small molecules with potential to prevent and/or reverse muscle atrophy. Unlike a conventional drug discovery approach, this strategy does not rely on a predefined molecular target but rather exploits the complexity of muscle atrophy to identify small molecules that counter the entire spectrum of pathological changes in atrophic muscle. We discuss how this strategy has been used to identify two natural compounds, ursolic acid and tomatidine, that reduce muscle atrophy and improve skeletal muscle function. SUMMARY Discovery strategies based on mRNA expression signatures can elucidate new approaches for preserving and restoring muscle mass and function.
Collapse
Affiliation(s)
- Christopher M. Adams
- Department of Internal Medicine, Department of Molecular Physiology and Biophysics, and the Fraternal Order of Eagles Diabetes Research Center, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City
- Iowa City Veterans Affairs Medical Center, Iowa City
- Emmyon, Inc., Coralville, Iowa, USA
| | - Scott M. Ebert
- Department of Internal Medicine, Department of Molecular Physiology and Biophysics, and the Fraternal Order of Eagles Diabetes Research Center, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City
- Emmyon, Inc., Coralville, Iowa, USA
| | - Michael C. Dyle
- Department of Internal Medicine, Department of Molecular Physiology and Biophysics, and the Fraternal Order of Eagles Diabetes Research Center, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City
| |
Collapse
|
200
|
Konishi M, Ebner N, von Haehling S, Anker SD, Springer J. Developing models for cachexia and their implications in drug discovery. Expert Opin Drug Discov 2015; 10:743-52. [DOI: 10.1517/17460441.2015.1041914] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|